WO2004075882A1 - Methods of preventing, treating and diagnosing disorders of protein aggregation - Google Patents

Methods of preventing, treating and diagnosing disorders of protein aggregation Download PDF

Info

Publication number
WO2004075882A1
WO2004075882A1 PCT/CA2004/000272 CA2004000272W WO2004075882A1 WO 2004075882 A1 WO2004075882 A1 WO 2004075882A1 CA 2004000272 W CA2004000272 W CA 2004000272W WO 2004075882 A1 WO2004075882 A1 WO 2004075882A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
disease
group
amyloid
dementia
Prior art date
Application number
PCT/CA2004/000272
Other languages
French (fr)
Inventor
Joanne Mclaurin
Original Assignee
Ellipsis Biotherapeutics Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to EP04715226A priority Critical patent/EP1608350B1/en
Priority to BRPI0407910-8A priority patent/BRPI0407910A/en
Priority to US10/547,286 priority patent/US8859628B2/en
Priority to AU2004216544A priority patent/AU2004216544B2/en
Priority to SI200431216T priority patent/SI1608350T1/en
Priority to JP2006501433A priority patent/JP4999453B2/en
Priority to CA2516563A priority patent/CA2516563C/en
Priority to DK04715226T priority patent/DK1608350T3/en
Application filed by Ellipsis Biotherapeutics Corporation filed Critical Ellipsis Biotherapeutics Corporation
Priority to DE602004021362T priority patent/DE602004021362D1/en
Priority to AT04715226T priority patent/ATE432694T1/en
Publication of WO2004075882A1 publication Critical patent/WO2004075882A1/en
Priority to IL170476A priority patent/IL170476A/en
Priority to IL220065A priority patent/IL220065A/en
Priority to US14/475,859 priority patent/US20150031776A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • A61K31/047Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates having two or more hydroxy groups, e.g. sorbitol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/0491Sugars, nucleosides, nucleotides, oligonucleotides, nucleic acids, e.g. DNA, RNA, nucleic acid aptamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/58Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances
    • G01N33/60Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances involving radioactive labelled substances
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • G01N33/6896Neurological disorders, e.g. Alzheimer's disease

Definitions

  • the invention relates to methods for treating Alzheimer's Disease and other amyloidoses; more particularly, it relates to methods for inhibiting and reducing amyloid fibril formation in therapeutic intervention in Alzheimer's disease and other amyloidoses.
  • Alzheimer's disease is characterized neuropathologically by amyloid deposits, neurofibrillary tangles, and selective neuronal loss.
  • the major component of the amyloid deposits is amyloid- ⁇ (A ⁇ ), a 39-43 residue peptide. Soluble forms of A ⁇ generated from cleavage of amyloid precursor protein are normal products of metabolism.
  • residues 1-42 (A ⁇ 42) in Alzheimer's disease was highlighted in the discovery that mutations in codon717 of the amyloid precursor protein gene, presenilin 1 and presenilin 2 genes result in an increased production of A ⁇ 42 over A ⁇ 1 -40.
  • a ⁇ -fibrils may be an early and intervenable step during the progression of AD. Formation of amyloid plaques, as well as neurotoxicity and inflammation may be direct or indirect consequences of the interaction of A with molecules containing sugar moieties.
  • a ⁇ interaction with glycosaminoglycans results in aggregation of A ⁇ possibly adding to their insolubility and plaque persistence.
  • Glycosaminoglycans have also been implicated in neuronal toxicity and microglial activation.
  • interaction with glycolipids such as gangliosides results in the stabilization and prevention of Ab fibril formation, as well as, the site of A ⁇ production.
  • the family of phosphatidylinositols results in acceleration of fibril formation.
  • the headgroup of phosphatidylinositol is myo- inositol, a naturally occurring simple sugar involved in lipid biosynthesis, signal transduction, and osmolarity control.
  • amyloid deposition i.e. organs or tissues lying outside the central nervous system
  • amyloid accumulation leading to organ dysfunction or failure.
  • systemic organs i.e. organs or tissues lying outside the central nervous system
  • amyloid accumulation leading to organ dysfunction or failure.
  • Alzheimer's disease and "systemic" amyloid diseases there is currently no cure or effective treatment, and the patient usually dies within 3 to 10 years from disease onset.
  • U.S. Patent No. 4,847,082 discloses the use of phytic acid, a phytate salt, an isomer or hydrolysate of phytic acid for the treatment of Alzheimer's disease. It also discloses that isomers of phytic acid or phytate salt comprise the hexakisphosphate myo-inositol, hexakisphosphate scyl -mosito , hexakisphosphate D-cAz>oinositol, hexakisphosphate L-c zz ' ro-inositol, hexakisphosphate /zeo-inositol and hexakisphosphate muco-mosito conformations.
  • Phytic acid is inositol- hexakisphosphate (IP6).
  • IP6 inositol- hexakisphosphate
  • U.S. Patent No. 5,112,814 discloses the use of phytic acid and isomers thereof for the treatment of Parkinson's disease. As is the case with U.S. Patent No.
  • AD Alzheimer's disease
  • Alzheimer's disease Much work in Alzheimer's disease has been accomplished, but little is conventionally known about compounds or agents for therapeutic regimes to arrest or reverse amyloid formation, deposition, accumulation and/or persistence that occurs in Alzheimer's disease and other amyloidoses.
  • New compounds or agents for therapeutic regimes to arrest or reverse amyloid formation, deposition, accumulation and/or persistence that occurs in Alzheimer's disease and other amyloidoses are therefore urgent needed.
  • the present invention provides a method of treating or preventing in a subject a condition of the central or peripheral nervous system or systemic organ associated with a disorder in protein folding or aggregation, or amyloid formation, deposition, accumulation, or persistence comprising administering to said subject a pharmaceutically effective amount of compound selected having the following structure:
  • each of Rmetabol R,,, R 2 , R 2 ,, R 3 , R 3 ,, R 4 , R 4 ., R 5 , R 5 ,, R g , and R 6 . is independently selected from the group of:
  • NHR7 wherein said R7 is selected from the group of hydrogen; C2- Cjo acyl and C ⁇ -CIQ alkyl;
  • RJQ is selected from the group of no group, hydrogen, C2-C10 acyl, C 1-C10 alkyl and SO3H;
  • the present invention also provides a method of preventing abnormal protein folding, abnormal protein aggregation, amyloid formation, deposition, accumulation, or persistence, or amyloid lipid interactions in a subject comprising administering to said subject a pharmaceutically effective amount of a compound having the following structure:
  • each of R privilege R r , R 2 , R 2 ., R 3 , R 3 ., R , R 4 -, R 5 , R 5 ., R 6 , and R 6 . is independently selected from the group of:
  • NHR7 wherein said R7 is selected from the group of hydrogen; C2- Cjo acyl and Ci-Cjo alkyl;
  • Rj Q wherein said Rj 0 is selected from the group of no group, hydrogen, C2-C10 acyl, C I-CJQ alkyl and SO3H;
  • each of R privilege R r , R 2 , R 2 ., R 3 , R 3 ., R 4 , R >, R 5 , R 5 ,, R 6 , and R 6 . is independently selected from the group of:
  • NHR7 wherein said R7 is selected from the group ofhydrogen; C2- C ⁇ 0 acyl and C ⁇ -C ⁇ Q alkyl;
  • Ri 0 is selected from the group of no group, hydrogen, C2-C10 acyl, C i-C ⁇ o alkyl and SO3H; (e) C5-C7 glycosyl;
  • SRj 1 wherein R ⁇ is selected from the group ofhydrogen, Cj-Cio alkyl and O3H; (h) C 1 -C ⁇ 0 alkyl optionally substituted with a substituent selected from the group of hydrogen, OR ⁇ o > NHR7, NR R9 and SRjj; and (i) C3 ⁇ Cg cycloalkyl optionally substituted with a substituent selected from the group ofhydrogen, ORJQ, NHR7, NRgRp and SRj ⁇ providing that the compound is riot myo-inositol.
  • the present invention also provides a method of diagnosing the presence of abnormally folded or aggregated protein and/or amyloid fibril or amyloid in a subject comprising: (a) administering to said subject a radioactive compound or compound tagged with a substance that emits a detectable signal in a quantity sufficient and under conditions to allow for the binding of said compound to the abnormally folded or aggregated protein and/or fibrils or amyloid, if present; and (b) detecting the radioactivity or the signal from the compound bound to the abnormally folded or aggregated protein and/or fibrils or amyloid, thus diagnosing the presence of abnormally folded or aggregated protein and/or amyloid fibril or amyloid in said subject, wherein said compound has the following structure:
  • each of R privilege R r , R 2 , R 2 ., R 3 , R 3 ,, R 4 , R 4 ,, R 5 , R 5 ,, R 6 , and R 6 is independently selected from the group of:
  • NHR7 wherein said R7 is selected from the group ofhydrogen; C2- CJO ac yl and Ci-CjQ alky
  • R Q is selected from the group of no group, hydrogen, C2-C10 acyl, C i-Cjo alkyl and SO3H; (e) C5-C7 glycosyl;
  • SRj 1 wherein R ⁇ is selected from the group ofhydrogen, Cj-Ci 0 alkyl and O3H;
  • CJ-CJO alkyl optionally substituted with a substituent selected from the group of hydrogen, ORJO NHR7 > NR R9 and SRj ⁇ and
  • C3-Cg cycloalkyl optionally substituted with a substituent selected from the group ofhydrogen, OR ⁇ o > NHR7, NRgR9 and SRj ⁇ providing that the compound is not /wyo-inositol.
  • the present invention further provides a method of diagnosing the presence of abnormally folded or aggregated protein and/or amyloid fibril or amyloid in a subject comprising: (a) collecting a sample from said subject; (b) contacting said sample with a radioactive compound or compound tagged with a substance that emits a detectable signal under conditions to allow the binding of said compound to the abnormally folded or aggregated protein and/or amyloid fibril or amyloid if present; and (c) detecting the radioactivity or the signal from the compound bound to the abnormally folded or aggregated protein and/or fibrils or amyloid, thus diagnosing the presence of abnormally folded or aggregated protein and/or amyloid fibril or amyloid in said subject, wherein said compound has the following structure:
  • each of R l5 R,. s R 2 , R 2 ., R 3 , R 3 , 5 R 4s R 4 ,, R 5 , R 5 , s R 6 , and R & is independently selected from the group of;
  • NHR7 wherein said R7 is selected from the group ofhydrogen; C2- C 10 acyl and C 1 -C ⁇ 0 alkyl;
  • Figure 1 A shows the structure of myo-, epi- and scyllo-mositol while Figures 1 B- 1 H show the spatial reference memory version of the Morris water maze test in TgCRND8 mice.
  • v ⁇ -inositol treatment did not alter cognitive function (IB).
  • Figures 2A-2I show at 6 months of age, the plaque burden and astrogliosis in TgCRND ⁇ treated with epi- and scy/Zo-inositol treated mice.
  • Control animals have a high plaque load and astrogliosis in the hippocampus (2A) and cerebral cortex (2B).
  • Higher magnification demonstrates that astrocytic activation is not only associated with plaque load (2C).
  • Epz ' -inositol treatment has a modest effect on amyloid burden with a decrease in astrogliosis (2D, 2 ⁇ and 2F).
  • Scy/Zo-inositol treatment significantly decreased amyloid burden and gliosis (2G, 2H, and 21).
  • FIGS 3A-3D show that the A ⁇ species, 1-42, 1-40 and 1-38, in control and treated TgCRND8 mice was indistinguishable (3A) as was the extent of APP processing (3B).
  • Vascular amyloid burden was quantitated on serial sagittal sections in treated and untreated TgCRND8 mice.
  • TgCRND ⁇ mice have a significant vascular amyloid burden that is associated with small and medium sized vessels, the load is decreased in scy/zO-inositol treated TgCRND8 mice (3A). Scyllo-inositol treatment significantly decreased the total vascular load in comparison to untreated and epi- inositol treated TgCRND8 mice (3C). Sc /Zo-inositol decreases plaque deposition as illustrated by the significant decrease in mean plaque size (3D).
  • Figure 4 shows the effect of water on the cognitive function of TgCRND8 and non-Tg mice using the spatial reference memory version of the Morris Water Maze in a three day trial paradigm.
  • Figure 5 shows the effect of scyllo-mos ⁇ .6 on the cognitive function of TgCRND8 and non-Tg mice using the spatial reference memory version of the Morris Water Maze in a three day trial paradigm.
  • Figure 6 shows the effect of epz ' -inositol on the cognitive function of TgCRND8 and non-Tg mice using the spatial reference memory version of the Morris Water Maze in a three day trial paradigm.
  • Figure 7 shows the effect of w oinositol on the cognitive function of TgCRND ⁇ and non-Tg mice using the spatial reference memory version of the Morris Water Maze in a three day trial paradigm.
  • Figure 8 shows the effect of scy/fo-inositol, epz ' -inositol and r ⁇ y ⁇ -inositol on the cognitive function of TgCRND8 (learning phase and memory test) and compared with wild type mice using the spatial reference memory version of the Morris Water Maze in a three-day trial paradigm.
  • Figure 9 shows the percentage of brain area covered with plaques in untreated TgCRND8 mice versus mice treated with scy/Zo-inositol, epz ' -inositol or myo-inositol.
  • Figures 10A and 10B show the survival rates of TgCRND ⁇ mice treated with water versus epz ' -inositol or myo-inositol (10A) or versus •s , cy// ⁇ -inositol (10B).
  • Figures 12A and B show the results of a spatial reference memory test in the treatment studies when performed in a 3-day trial paradigm.
  • Figures 13 A and B show A ⁇ levels within the CNS after administration of various doses of scy//o-inositol were administered once daily for one month to five month old TgCRNDS mice. Soluble A ⁇ 42 levels were decreased at all doses and were significantly different from untreated controls (A). In contrast, insoluble A ⁇ 42 was not significantly different under all conditions (B). Vertical bars represent S.E.M.
  • Figure 15 shows the cognitive performance of 6-month old ⁇ //o-inositol- treated TgCRND ⁇ mice compared with that of their non-transgenic littermates.
  • Figures 16A-D show that at 2 months of age, the plaque burden in TgPSl x APP mice is decreased in scy/Zo-inositol treated mice.
  • Control animals have a high plaque load in the hippocampus (A) and cerebral cortex (B). Scy/zo-inositol treatment significantly decreased amyloid burden (C, D). Plaque burden identified using anti- A ⁇ antibody (brown). Scale Bar 300 ⁇ m.
  • Figures 17A-C show the quantification of the plaque burden in TgPSlxAPP mice after -? ⁇ y//o-inositol treatment.
  • the percent brain area covered in plaques (A), mean plaque size (B) and plaque count (C) were significantly reduced.
  • Vertical bars are S.E.M. DETAILED DESCRIPTION OF THE INVENTION
  • the present invention discloses novel, unpredictable and unexpected properties of certain inositol stereoisomers in relation to the treatment of amyloid- related disorders such as Alzheimer's Disease. It has been surprisingly discovered that certain stereoisomers of inositol and related compounds block A ⁇ -induced progressive cognitive decline and cerebral amyloid plaque pathology, and improve survival when given to a transgenic mouse model of human Alzheimer Disease during the nascent phase of A ⁇ deposition.
  • inositol stereoisomers e.g. epi- and scy// ⁇ -inositols
  • the present invention describes the unexpected results that scy/Zo-inositol inhibits already established cerebral amyloid deposition, and does so in the living brain. This is not implied by the previously published in vitro data which considered only certain neuronal cell types in culture, not the complex tissues of the living brain, and only suggested that inositols might inhibit de novo aggregation, thereby having no relevance to established disease.
  • the compounds of the present invention are 1,2,3,4,5,6- cyclohexanehexols, more preferably selected from the group of cis-, epi-, allo-, muco-, neo- , scyllo-, D-chiro- and L-cAzro-inositols.
  • these compounds are 1,2,3,4,5-cyclohexanepentols (quercitols), more preferably selected from the group of epi-, vibo-, scyllo-, allo-, talo-, gala-, cis-, muco-, neo-, pr ⁇ t ⁇ -quercitols and enantiomers thereof.
  • quercitols 1,2,3,4,5-cyclohexanepentols
  • these compounds are selected from the group of a cyclohexanetetrol, a cyclohexanetriol, stereoisomer of cyclohexanetetrol, stereoisimer of cyclohexanetriol, enantiomer of cyclohexanetetrol, and enantiomer of cyclohexanetriol.
  • These compounds may also be compound is pentahydxycyclohexanones or stereoisomers or enantiomers thereof. Yet again preferably, these compounds are inosose compounds selected from the group of scyZZ ⁇ -inosose, L-c/ ⁇ z ' r ⁇ -inosose-1 and L-epz ' -inosose.
  • these compounds are trihydroxyxcyclohexanones, or stereoisomers or enantiomers thereof. More preferably, (-)-l- e ⁇ y-scyllo-inosose.
  • these compounds are pentahydxycyclohexanones (inosose), or stereoisomers or enantiomers thereof, more preferably selected from the group of .scy/Z ⁇ -inosose, L-c/zz ' r ⁇ -inosose-l and L-epz ' -inosose.
  • these compounds are trihydroxyxcyclohexanones or stereoisomers or enantiomers thereof such as (-)-l-deoxy-scyllo-mosose.
  • these compounds are O-monomethyl-cyclohexanehexols or stereoisomers or enantiomers thereof, more preferably selected from the group of D- pinitol, L-quebrachitol and D-bornesitol.
  • these compounds may be selected from the group of , monoaminocyclohexanepentols (inosamines), diaminocyclohexanetetrols (inosadiamines), diaminocyclohexanetriols, stereoisomers thereof, and enantiomers thereof, and pharmaceutically acceptable salts thereof such as L-ne ⁇ -inosamine, D,L- epz ' -inosamine-2, streptamine and deoxystreptamine.
  • these compounds are monomercapto-cyclohexanepentols or stereoisomers or enantiomers thereof, more preferably 1L-1 - eo y-1 -mercapto-8- O-methyl-chiro-inositol.
  • the most preferred compounds of the present invention are ⁇ ZZ ⁇ -inositol and scyZZo-inositol, with scyZZ ⁇ -inositol being the most preferred.
  • the inositol stereoisomers of the present invention exclude zwy ⁇ -inositol and may also exclude epz ' -inositol.
  • these compounds are found to be useful in treating or preventing in a subject a condition of the central or peripheral nervous system or systemic organ associated with a disorder in protein folding or aggregation, or amyloid formation, deposition, accumulation, or persistence.
  • These compounds are also found to be useful in preventing abnormal protein folding, abnormal protein aggregation, amyloid formation, deposition, accumulation, or persistence, or amyloid lipid interactions as well as causing the dissociation of abnormally aggregated proteins and/or dissolving or disrupting pre-formed or pre-deposited amyloid fibril or amyloid in a subject.
  • the condition of the central or peripheral nervous system or systemic organ results in the deposition of proteins, protein fragments and peptides in beta-pleated sheats and/or fibrils and/or aggregates. More preferably, the condition of the central or peripheral nervous system or systemic organ is selected from the group of: Alzheimer's disease, presenile and senile forms; amyloid angiopathy; mild cognitive impairment; Alzheimer's disease-related dementia; tauopathy; ⁇ - synucleinopathy; Parkinson's disease; Amyotrophic Lateral Sclerosis; motor neuron Disease; Spastic paraplagia; Huntington's Disease, spinocerebellar ataxia, Freidrich's Ataxia; neurodegenerative diseases associated with intracellular and/or intraneuronal aggregates of proteins with polyglutamine, polyalanine or other repeats arising from pathological expansions of tri- or tetra-nucleotide elements within corresponding genes; cerebro vascular diseases; Down's syndrome; head trauma with post-traumatic accumulation of
  • Alzheimer dementia and tauopathy selected from the group of argyrophilic grain dementia, corticobasal degeneration, dementia pugilistica, diffuse neurofibrillary tangles with calcification, frontotemporal dementia with parkinsonism, Prion-related disease, Hallervorden-Spatz disease, myotonic dystrophy, Niemann-Pick disease type C, non-Guamanian Motor Neuron disease with neurofibrillary tangles, Pick's disease, postencephalitic parkinsonism, prion protein cerebral amyloid angiopathy, progressive subcortical gliosis, progressive supranuclear palsy, subacute sclerosing panencephalitis, and tangle only dementia.
  • the ⁇ -synucleinopathy is selected from the group of dementia with Lewy bodies, multiple system atrophy with glial cytoplasmie inclusions, Shy- Drager syndrome, striatonigral degeneration, olivopontocerebellar atrophy, neurodegeneration with brain iron accumulation type I, olfactory dysfunction, and amyotrophic lateral sclerosis.
  • the Motor Neuron Disease is associated with filaments and aggregates of neuro filament and/or superoxide dismutase proteins
  • the Spastic paraplegia is associated with defective function of chaperones arid/or triple A proteins
  • the spinocerebellar ataxia is DRPLA or Machado- Joseph Disease.
  • the Prion related disease is selected from the group of Creutzfeldt- Jakob disease, Gerstmann-Straussler-Scheinker disease, and variant Creutzfeldt-Jakob disease and the Amyloid Polyneuropathy is Senile amyloid polyneuropathy or Systemic Amyloidosis.
  • the condition of the central or peripheral nervous system or systemic organ is Parkinson's disease including familial and non-familial types. Most preferably, said condition of the central or peripheral nervous system or systemic organ is Alzheimer's disease.
  • the compound is administered to the subject at a dose of about 1 mg to about 1 g per kg, preferably 1 mg to about 200 mg per kg, more preferably about 10 mg to about 100 mg per kg and most preferably about 30 mg to 70 mg per kg of the weight of said subject.
  • the administration can be accomplished by a variety of ways such as orally (oral pill, oral liquid or suspension), intravenously, intramuscularly, intraperitoneally, intraderrnally, transcutaneously, subcutaneously, intranasally, sublingually, by rectal suppository or inhalation, with the oral administration being the most preferred.
  • the administration of the compounds of the present invention can be undertaken at various intervals such as once a day, twice per day, once per week, once a month or continuously.
  • the compounds of the present invention are administered in combination with other treatments such as beta-secretase inhibitors, gamma-secretase inhibitors (APP-specific or non-specific), epsilon-secretase inhibitors (APP-specific or non-specific), other inhibitors of beta-sheet aggregation/fibrillogenesis/ADDL formation (e.g. Alzhemed), NMDA antagonists (e.g. memantine), non-steroidal anti- inflammatory compounds (e.g. Ibuprofen, Celebrex), anti-oxidants (e.g. Vitamin E), hormones (e.g. estrogens), nutrients and food supplements (e.g.
  • other treatments such as beta-secretase inhibitors, gamma-secretase inhibitors (APP-specific or non-specific), epsilon-secretase inhibitors (APP-specific or non-specific), other inhibitors of beta-sheet aggregation/fibrillogenesis/ADDL formation (e.g. Alzhemed
  • Gingko biloba Gingko biloba
  • acetylcholinesterase inhibitors e.g. donezepil
  • muscarinic agonists e.g. AF102B (Cevimeline, EVOXAC), AF150(S), and AF267B
  • anti-psychotics e.g. haloperidol, clozapine, olanzapine
  • anti-depressants including tricyclics and serotonin reuptake inhibitors (e.g.
  • Sertraline and Citalopram Hbr gene therapy and/or drug based approaches to upregulate neprilysin (an enzyme which degrades A ⁇ ); gene therapy and/or drug based approaches to upregulate insulin degrading enzyme (an enzyme which degrades A ⁇ ), vaccines, immunotherapeutics and antibodies to A ⁇ (e.g. ELAN AN- 1792), statins and other cholesterol lowering drugs (e.g. Lovastatin and Simvastatin), stem cell and other cell-based therapies, inhibitors of kinases (CDK5, GSK3 ⁇ , GSK3 ⁇ ) that phosphorylate TAU protein (e.g. Lithium chloride), or inhibitors of kinases that modulate A ⁇ production (GSK3 ⁇ , GSK3 ⁇ , Rho/ROCK kinases) (e.g. lithium Chloride and Ibuprofen).
  • CDK5 inhibitors of kinases
  • GSK3 ⁇ , GSK3 ⁇ that phosphorylate TAU protein
  • the compounds of the present invention are also useful in diagnosing the presence of abnormally folded or aggregated protein and or amyloid fibril or amyloid in a subject using a method that comprises administering to said subject a radioactive compound or compound tagged with a substance that emits a detectable signal in a quantity sufficient and under conditions to allow for the binding of said compound to the abnormally folded or aggregated protein and/or fibrils or amyloid, if present; and detecting the radioactivity or the signal from the compound bound to the abnormally folded or aggregated protein and/or fibrils or amyloid, thus diagnosing the presence of abnormally folded or aggregated protein and/or amyloid fibril or amyloid.
  • a sample suspected of containing abnormally folded or aggregated protein and or amyloid fibril or amyloid is collected from a subject and is contacted with a radioactive compound or compound tagged with a substance that emits a detectable signal under conditions to allow the binding of said compound to the abnormally folded or aggregated protein and/or amyloid fibril or amyloid if present; and thereafter detect the radioactivity or the signal from the compound bound to the abnormally folded or aggregated protein and/or fibrils or amyloid, thus diagnosing the presence of abnormally folded or aggregated. protein and/or amyloid fibril or amyloid in said subject.
  • said detectable signal is a fluorescent or an enzyme-linked i ⁇ rmunosorbent assay signal and said sample is whole blood (including all cellular constituents) or plasma.
  • said detectable signal is a fluorescent or an enzyme-linked i ⁇ rmunosorbent assay signal and said sample is whole blood (including all cellular constituents) or plasma.
  • the compounds of the present invention can abrogate the cerebral accumulation of A ⁇ , the deposition of cerebral amyloid plaques, and cognitive decline in a transgenic mouse model of Alzheimer Disease when given during the "late presymptomatic" phase, prior to the onset of overt cognitive deficits and amyloid neuropathology in these mice. Furthermore, even when these compounds are given after the onset of cognitive deficits and amyloid plaque neuropathology, they can effectively reverse the amyloid deposition and neuropathology. Importantly, the mechanism of action of these compounds follows a rational design based upon their capacity to modulate the assembly of A ⁇ monomers into neurotoxic oligomers and/or protofibrils.
  • TgCRND8 mice are a robust murine model of Alzheimer's disease as described by Janus et al. (Nature 408:979-982 (2000). They express a human amyloid precursor protein (APP695) transgene under the regulation of the Syrian hamster prion promoter on a C3H/B6 outbred background. The human APP695 transgene bears two mutations that cause AD in humans (K670N/M671L and V717F).
  • APP695 human amyloid precursor protein
  • TgCRND ⁇ mice have progressive spatial learning deficits that are accompanied by rising cerebral A ⁇ levels and by increasing number of cerebral extracellular amyloid plaques that are similar to those seen in the brains of humans with AD (C. Janus et al., Nature 408:979-982 (2000)).
  • mice and non-transgenic littermates were either untreated, or were given a compound of the present invention as indicated below at 30mg/day/mouse beginning at age of about 6 weeks. The mice were followed for outcome measures cognitive function, brain A ⁇ levels, brain pathology, and survival at 4 months and 6 months of age.
  • mice Experimental groups of TgCRND ⁇ mice were fed myo-, epi- and sc ZZo-inositol at 30 mg/mouse/day. Two cohorts entered the study at 6 weeks of age and outcomes were analyzed at 4- and 6-months of age. The body weight, coat characteristics and in cage behavior was monitored. All experiments were performed according to the Canadian Council on Animal Care guidelines. Behavioral tests - After non-spatial pre-training, mice underwent place discrimination training for 5 days with 4 trials per day. Behavioral data was analyzed using a mixed model of factorial analysis of variance (ANOVA) with drug or genotype and training sessions as repeated measure factors.
  • ANOVA factorial analysis of variance
  • Amyloid plaque burden was assessed using Leco JA-3001 image analysis software interfaced with Leica microscope and Hitachi KP-M1U CCD video camera. Vascular burden was analyzed similarly and a dissector was used to measure the diameter of affected vessels.
  • Plasma and Cerebral A ⁇ Content - Hemi-brain samples were homogenized in a buffered sucrose solution, followed by either 0.4% diethylamine/lOOmM NaCI for soluble A ⁇ levels or cold formic acid for the isolation of total A ⁇ . After neutralization the samples were diluted and analyzed for A ⁇ 40 and A ⁇ 42 using commercially available kits (BIOSOURCE International). Each hemisphere was analyzed in triplicate with the mean ⁇ SEM reported. Western blot analyses were performed on all fractions using urea gels for A ⁇ species analyses. A ⁇ was detected using 6E10 (BIOSOURCE International) and Enhanced Chemiluminenscence (Amersham).
  • Sections were irnrnunolabelled for astrocytes with anti-rat GFAP IgG 2a (Dako; diluted 1 :50) and for microglia with anti-rat CD68 IgG2b (Dako; 1 :50).
  • Digital images were captured using a Coolsnap digital camera (Photometries, Tuscon, Arizona) mounted to a Zeiss Axioscope 2 Plus microscope. Images were analysed using Openlab 3.08 imaging software (Improvision, Lexington MA).
  • Example 2 Prevention of cognitive deficits
  • the cognitive function of TgCRND ⁇ mice was assessed using the spatial reference memory version of the Morris Water Maze using a five-day trial paradigm ( Figures 1C-1H).
  • ANOVA analysis of variance
  • TgCRND ⁇ mice treated with either epi- or _? ⁇ y// ⁇ -inositol performed significantly better than untreated TgCRND ⁇ mice (p ⁇ 0.02; Figs. 1C and D).
  • epz ' -inositol treated TgCRND ⁇ mice had a slightly slower learning curve during the first three days of training.
  • epz ' -inositol treated TgCRNDS mice were not statistically different from their non-Tg littermates (Fig. 2E).
  • scyllo- inositol treated TgCRND ⁇ mice were indistinguishable from non-Tg littermates on all days.
  • TgCRND ⁇ mice have high plasma A ⁇ concentrations at 4-months of age and remain constant at 6 months of age even though CNS plaque load is still rising at 6-months of age (Table 1). Neither epz ' -inositol nor sey/Zo-inositol treatment had any effect on plasma A ⁇ levels in comparison to untreated TgCRNDS mice (p-0.89).
  • Astroglial and microglial reactions are neuropathological features both of human AD and of all amyloid mouse models (Irizarry et al., J Neuropathol Exp Neurol 56 , 965, 1997; K. D. Bornemann et al. Ann N Y Acad Sci. 90 ⁇ , 260, 2000). Therefore, the effect of epi- and scy/Z ⁇ -inositol treatment was investigated on astrogliosis and microgliosis in the brains of TgCRND8 mice ( Figures 3A-3D). Serial sagittal sections were stained with the astrocytic marker glial fibrillary acidic protein (GFAP) and quantitated for percent brain area covered by astrogliosis.
  • GFAP astrocytic marker glial fibrillary acidic protein
  • TgCRND ⁇ mice have a high basal astrogliosis at 4-months of age (0.459 ⁇ 0.04 ⁇ %), which increases slightly by 6-months of age (0.5 ⁇ 4 ⁇ 0.0 ⁇ 9%), and which is not restricted to plaque areas ( Figures 2A-C).
  • Microglial activation was also significantly attenuated in scy// ⁇ -inositol treated TgCRND ⁇ mice (0.20 ⁇ 0.008% brain area) when compared to age- and sex-matched untreated TgCRNDS mice (0.31 ⁇ 0.01%; p ⁇ .001).
  • Alzheimer's disease is characterized by the presence of both parenchymal and vascular amyloid deposits.
  • TgCRND ⁇ mice approximately 0.03% of the brain area is associated with vascular amyloid.
  • No difference could be detected in the vascular amyloid burden after epz ' -inositol treatment at 6 months of age ( Figure 3C).
  • Treatment with ⁇ -inositol did not affect overall survival significantly ( Figure 10A).
  • treatment of wild type mice with sc ZZo-inositol had no effect either on survival or on other parameters such as weight, fur condition or cage behavior.
  • Example 7 Treatment and Reversal of amyloid deposition
  • scyllo-inositol inhibits both parenchymal and cerebrovascular amyloid deposition and results in improved survival and cognitive function in the TgCRND8 mouse model of Alzheimer disease.
  • most Alzheimer's disease patients will likely seek treatment only once symptomatic, and when A ⁇ oligomerization, deposition, toxicity and plaque formation are already well advanced within the CNS.
  • a pilot study was therefore initiated on 5 month old TgCRND ⁇ mice. These mice have significant A ⁇ and plaque burdens that are comparable to those in the brain of humans with AD.
  • mice mice were fed myo-, epi- and scyllo- inositol at 30 mg/mouse/day. A cohort entered the study at 5 months of age and outcomes were analyzed at 6-months of age. The body weight, coat characteristics and in cage behavior was monitored. All experiments were performed according to the Canadian Council on Animal Care guidelines.
  • Behavioral Test - Reversal Study Mice entered the Morris water maze test with a hidden platform on day one without pretraining. Mice were tested for 3 days with six trials per day. On the fourth day, the platform was removed from the pool and each mouse received one 30-s swim probe trial. On the last day the animals underwent a cue test in order to evaluate swimming ability, eye sight and general cognition. The cue test is composed at the platform being placed in a different quadrant than that used for testing and is tagged with a flag. Animals are allowed 60 s to find the platform. Animals that do not find the platform are not used in the final analyses of spatial memory. Behavioural data was analysed using a mixed model of factorial analysis of variance (ANOVA) with drug or genotype and training sessions as repeated measure factors.
  • ANOVA factorial analysis of variance
  • Plasma and Cerebral A ⁇ Content - Hemi-brain samples were homogenized in a buffered sucrose solution, followed by either 0.4% diethylamine/lOOmM NaCI for soluble A ⁇ levels or cold formic acid for the isolation of total A ⁇ . After neutralization the samples were diluted and analyzed for A ⁇ 40 and A ⁇ 42 using commercially available kits (BIOSOURCE International). Each hemisphere was analyzed in triplicate with the mean ⁇ SEM reported.
  • ANOVA analysis of variance
  • TgCRND8 mice were significantly impaired in comparison to wild type littermates (Figure 4).
  • Example 10 Efficacy of ⁇ /fo-inositol for the treatment of disease bearing TgCRND8 mice.
  • TgCRND ⁇ mice were either treated for 2 days with allo- inositol, or were untreated.
  • the dosage and oral administration of compounds, and the behavioral and neurochemical assays were the same as those employed in the above treatment experiments.
  • Cerebral A ⁇ levels were analyzed for treatment versus untreated TgCRND ⁇ mice to determine whether improved behavior could be correlated with changes in A ⁇ (Table 4).
  • ⁇ ZZo-inositol treatment reduced soluble A ⁇ 42 (20% reduction, p ⁇ 0.05) an effect similar to that seen for scyllo- inositol.
  • y ⁇ ZZ ⁇ -inositol did not significantly alter insoluble A ⁇ 42 or A ⁇ 40 (soluble and insoluble pools).
  • One possible explanation for the decrease in A ⁇ 42 is clearance of A ⁇ 42 in the periphery with a subsequent increase in plasma A ⁇ 42.
  • Tg PSl x APP mice are an enhanced model of Alzheimer's disease which express a mutant human PSl transgene encoding two familial mutations (M146L and L2 ⁇ 6V) in conjunction with the human APP transgene encoding the Indiana and Swedish familial mutations. These animals develop robust expression of cerebral A ⁇ levels and amyloid deposition by 30-45 days of age. In a prophylactic trial, TgPSlxAPP mice were treated with -fcy/Z ⁇ -inositol from weaning and were assessed for effects on neuropathology at 2 months of age ( Figures 16 and 17).
  • scyllo- inositol treated TgPSlxAPP mice displayed a significant decrease in all measures of plaque burden at 2 months of age.
  • TgCRND ⁇ mice were treated with a simple sugar of similar molecular weight, mannitol. At 6 months of age, mannitol treated TgCRND ⁇ mice were indistinguishable from untreated TgCRND ⁇ mice (Fig. 11 A) and were significantly different from mannitol treated non-Tg littermates (Fig. 1 IB). Mannitol had no effect on the behaviour of non-Tg mice, since mannitol treated non-Tg mice were indistinguishable from untreated non-Tg mice.

Abstract

Disclosed are methods of preventing, treating, or diagnosing in a subject a disorder in protein folding or aggregation., or amyloid formation., deposition, accumulation, or persistence consisting of administering to said subject a pharmaceutically effective amount of inositol stereoisomers, enantiomers or derivatives thereof.

Description

METHODS OF PREVENTING. TREATING AND DIAGNOSING DISORDERS OF PROTEIN AGGREGATION
RELATED APPLICATIONS
This application claims the priority of U.S. Provisional Application Serial Nos. 60/451 ,363, 60/520,958 and 60/523,534, filed February 27, 2003, November 17, 2003 and November 19, 2003, respectively.
FIELD OF THE INVENTION
The invention relates to methods for treating Alzheimer's Disease and other amyloidoses; more particularly, it relates to methods for inhibiting and reducing amyloid fibril formation in therapeutic intervention in Alzheimer's disease and other amyloidoses.
DESCRIPTION OF THERELATED ART
Alzheimer's disease is characterized neuropathologically by amyloid deposits, neurofibrillary tangles, and selective neuronal loss. The major component of the amyloid deposits is amyloid- β(Aβ), a 39-43 residue peptide. Soluble forms of Aβ generated from cleavage of amyloid precursor protein are normal products of metabolism. The importance of residues 1-42 (Aβ42) in Alzheimer's disease was highlighted in the discovery that mutations in codon717 of the amyloid precursor protein gene, presenilin 1 and presenilin 2 genes result in an increased production of Aβ42 over Aβ 1 -40. These results in conjunction with the presence of Aβ 42 in both mature plaques and diffuse amyloid lead to the hypothesis that this more amyloidogenic species may be the critical element in plaque formation. This hypothesis was supported by the fact that Aβ42 deposition precedes that of Aβ40 in Down's syndrome in PS1 mutations and in hereditary cerebral hemorrhage with amyloidosis. Many in vitro studies have demonstrated that Aβ can be neurotoxic or enhance the susceptibility of neurons to excitotoxic, metabolic, or oxi dative insults. Initially it was thought that only the fibrillar form of A was toxic to neurons but more thorough characterization of Aβ structures demonstrated that dimers and small aggregates of Aβ are also neurotoxic. These data suggested that prevention of Aβ oligomerization would be a likely strategy to prevent AD-related neurodegeneration. Several studies have demonstrated that in vitro Aβ-induced neurotoxicity can be ablated by compounds that can increase neuronal resistance by targeting cellular pathways involved in apoptosis, block downstream pathways after Aβ induction of destructive routes, or block Aβ oligomerization and ultimately fibril formation. The site at which Aβ acts to induce neurotoxicity has yet to be elucidated but its toxic effects have been blocked by a variety of disparate agents.
Docking of Aβ-fibrils to neuronal and glial cell membranes may be an early and intervenable step during the progression of AD. Formation of amyloid plaques, as well as neurotoxicity and inflammation may be direct or indirect consequences of the interaction of A with molecules containing sugar moieties. Previous studies have demonstrated that Aβ interaction with glycosaminoglycans results in aggregation of Aβ possibly adding to their insolubility and plaque persistence. Glycosaminoglycans have also been implicated in neuronal toxicity and microglial activation. Alternatively, interaction with glycolipids such as gangliosides results in the stabilization and prevention of Ab fibril formation, as well as, the site of Aβ production. The family of phosphatidylinositols, on the other hand, results in acceleration of fibril formation. The headgroup of phosphatidylinositol is myo- inositol, a naturally occurring simple sugar involved in lipid biosynthesis, signal transduction, and osmolarity control.
It is also noteworthy that a variety of other human diseases also demonstrate amyloid deposition and usually involve systemic organs (i.e. organs or tissues lying outside the central nervous system), with the amyloid accumulation leading to organ dysfunction or failure. In Alzheimer's disease and "systemic" amyloid diseases, there is currently no cure or effective treatment, and the patient usually dies within 3 to 10 years from disease onset.
U.S. Patent No. 4,847,082 discloses the use of phytic acid, a phytate salt, an isomer or hydrolysate of phytic acid for the treatment of Alzheimer's disease. It also discloses that isomers of phytic acid or phytate salt comprise the hexakisphosphate myo-inositol, hexakisphosphate scyl -mosito , hexakisphosphate D-cAz>oinositol, hexakisphosphate L-c zz'ro-inositol, hexakisphosphate /zeo-inositol and hexakisphosphate muco-mosito conformations. Phytic acid is inositol- hexakisphosphate (IP6). U.S. Patent No. 5,112,814 discloses the use of phytic acid and isomers thereof for the treatment of Parkinson's disease. As is the case with U.S. Patent No.
4,847,082, the phytic acid isomers disclosed in this patent retain the six phosphate groups on the six-carbon inositol sugar.
It is noteworthy that in subsequent publications, the ability of inositol- monophosphate, inositol-1 ,4-bisphosphate and inositol-1 ,4,5-triphosphate to inhibit amyloid-beta peptide fibrillogenesis were investigated and found not to be effective
(J. Mol. Biol. 278:183-194, 1998).
Barak et al. disclose the use of inositol for the treatment of Alzheimer's
Disease (AD). (Prog Neuro-psychoparmacol & Biol Psychiat. 20:729-735, 2000). However, this reference does not disclose the use of inositol isomers. Patients treated with inositol did not show any significant differences in overall cognitive function scores (CAMCOG index) between inositol and placebo (dextrose) in AD patients while two specific subscales of the CAMCOG index did show significant improvement (orientation and language). Levine J. reviews the above Barak et al. paper and specifically states that inositol treatment is not beneficial in AD or ECT-induced cognitive impairment (Eur
Neuropsychoparm. 1997; 7,147-155, 1997).
Colodny L, et al. suggests further studies for the usefulness of inositol in
Alzheimer's disease by referring to the above Barak et al. paper and therefore does not disclose or suggest such use for inositol isomers (Altern Med Rev 3(6):432-47, 1998). McLaurin et al. disclosed that m σ-inositol stabilizes a small micelle of Aβ42 (J. Mol. Biol. 278, 183-194, 1998). In addition, McLaurin et al. disclose that epi- and scyllo- but not cAzro-inositol were able to induce a structural transition from random to β-strucrare in Aβ42 (J Biol Chem. Jun 16; 275(24): 18495-502, 2000; and J Struct Biol 130:259-270, 2000). Alternatively, none of the stereoisomers were able to induce a structural transition in Aβ40. Electron microscopy showed that inositol stabilizes small aggregates of Aβ42. These references also disclose that inositol-Aβ interactions result in a complex that is non-toxic to nerve growth factor-differentiated PC- 12 cells and primary human neuronal cultures.
Much work in Alzheimer's disease has been accomplished, but little is conventionally known about compounds or agents for therapeutic regimes to arrest or reverse amyloid formation, deposition, accumulation and/or persistence that occurs in Alzheimer's disease and other amyloidoses.
New compounds or agents for therapeutic regimes to arrest or reverse amyloid formation, deposition, accumulation and/or persistence that occurs in Alzheimer's disease and other amyloidoses are therefore desperately needed.
SUMMARY OF THE INVENTION
The present invention provides a method of treating or preventing in a subject a condition of the central or peripheral nervous system or systemic organ associated with a disorder in protein folding or aggregation, or amyloid formation, deposition, accumulation, or persistence comprising administering to said subject a pharmaceutically effective amount of compound selected having the following structure:
Figure imgf000005_0001
wherein each of R„ R,,, R2, R2,, R3, R3,, R4, R4., R5, R5,, Rg, and R6. is independently selected from the group of:
(a) hydrogen atom;
(b) NHR7, wherein said R7 is selected from the group of hydrogen; C2- Cjo acyl and C\-CIQ alkyl;
(c) NRg 9, wherein said Rg is C2-C10 acyl or C\-C Q alkyl and said R9 is C2-C10 acyl or
Figure imgf000006_0001
alkyl;
(d) ORιo> wherein said RJQ is selected from the group of no group, hydrogen, C2-C10 acyl, C 1-C10 alkyl and SO3H;
(e) C5-C7 glycosyl;
(f) C3-C8 cycloalkyl optionally substituted with a substituent selected from the group of hydrogen, OH, NH2, SH, OSO3H and OPO3H2;
(g) SRJ I, wherein Ru is selected from the group ofhydrogen, Cj-Cio alkyl and O3H;
(h) C 1 -C 10 alkyl optionally substituted with a substituent selected from the group ofhydrogen, OR} Q, NHR , NRgRα. and SRj \ ; and
(i) C3-C cycloalkyl optionally substituted with a substituent selected from the group ofhydrogen, OR]Q3 NHR7, NRgR9 and SRj \ providing that the compound is not mvo-inositol.
The present invention also provides a method of preventing abnormal protein folding, abnormal protein aggregation, amyloid formation, deposition, accumulation, or persistence, or amyloid lipid interactions in a subject comprising administering to said subject a pharmaceutically effective amount of a compound having the following structure:
Figure imgf000006_0002
wherein each of R„ Rr, R2, R2., R3, R3., R , R4-, R5, R5., R6, and R6. is independently selected from the group of:
(a) hydrogen atom;
(b) NHR7, wherein said R7 is selected from the group of hydrogen; C2- Cjo acyl and Ci-Cjo alkyl;
(c) NRg , wherein said Rg is C2~Cι 0 acyl or Cj-Cio a'^ ^ said R9 is C2-C10 acyl or CJ-CIQ alkyl;
(d) ORj Q, wherein said Rj 0 is selected from the group of no group, hydrogen, C2-C10 acyl, C I-CJQ alkyl and SO3H;
(e) C5-C7 glycosyl;
(f) C3~Cg cycloalkyl optionally substituted with a substituent selected from the group ofhydrogen, OH, NH2, SH, OSO3H and OPO3H2;
(g) SRi 1 , wherein Rj \ is selected from the group ofhydrogen, CI-CJO alkyl and O3H;
(h) C 1 -C 10 alkyl optionally substituted with a substituent selected from the group ofhydrogen, O jo* NHR7> N gR9 and SR^; and
(i) C3-C cycloalkyl optionally substituted with a substituent selected from the group ofhydrogen, ORJQ, NHR7, NRgRp and SRj ] providing that the compound is not røvo-inositol.
The present invention further provides a method of causing the dissociation of abnormally aggregated proteins and or dissolving or disrupting pre-formed or pre- deposited amyloid fibril or amyloid in a subject comprising administering to said subject a pharmaceutically effective amount of a compound having the following structure:
Figure imgf000007_0001
wherein each of R„ Rr, R2, R2., R3, R3., R4, R >, R5, R5 ,, R6, and R6. is independently selected from the group of:
(a) hydrogen atom;
(b) NHR7, wherein said R7 is selected from the group ofhydrogen; C2- C\ 0 acyl and C\ -C\ Q alkyl;
(c) NR R9, wherein said Rg is C2-C10 acyl or C -CIQ alkyl and said R9 is C2-C10 acyl or C^-Cio alkyl;
(d) OR] 0, wherein said Ri 0 is selected from the group of no group, hydrogen, C2-C10 acyl, C i-C^o alkyl and SO3H; (e) C5-C7 glycosyl;
(f) C3~C cycloalkyl optionally substituted with a substituent selected from the group ofhydrogen, OH, NH2, SH, OSO3H and OPO3H2;
(g) SRj 1 , wherein R\ is selected from the group ofhydrogen, Cj-Cio alkyl and O3H; (h) C 1 -C \ 0 alkyl optionally substituted with a substituent selected from the group of hydrogen, ORιo> NHR7, NR R9 and SRjj; and (i) C3~Cg cycloalkyl optionally substituted with a substituent selected from the group ofhydrogen, ORJQ, NHR7, NRgRp and SRj \ providing that the compound is riot myo-inositol. The present invention also provides a method of diagnosing the presence of abnormally folded or aggregated protein and/or amyloid fibril or amyloid in a subject comprising: (a) administering to said subject a radioactive compound or compound tagged with a substance that emits a detectable signal in a quantity sufficient and under conditions to allow for the binding of said compound to the abnormally folded or aggregated protein and/or fibrils or amyloid, if present; and (b) detecting the radioactivity or the signal from the compound bound to the abnormally folded or aggregated protein and/or fibrils or amyloid, thus diagnosing the presence of abnormally folded or aggregated protein and/or amyloid fibril or amyloid in said subject, wherein said compound has the following structure:
Figure imgf000009_0001
wherein each of R„ Rr, R2, R2., R3, R3,, R4, R4,, R5, R5 ,, R6, and R6, is independently selected from the group of:
(a) hydrogen atom;
(b) NHR7, wherein said R7 is selected from the group ofhydrogen; C2- CJO acyl and Ci-CjQ alky
(c) NRgR9, wherein said Rg is C2-C10 acyl or C^-Ci 0 alkyl and said R9 is C2-C10 acyl or C1 -C1 Q alkyl;
(d) ORJOJ wherein said R Q is selected from the group of no group, hydrogen, C2-C10 acyl, C i-Cjo alkyl and SO3H; (e) C5-C7 glycosyl;
(f) C3~Cg cycloalkyl optionally substituted with a substituent selected from the group ofhydrogen, OH, NH2, SH, OSO3H and OPO3H2;
(g) SRj 1 , wherein R \ is selected from the group ofhydrogen, Cj-Ci 0 alkyl and O3H; (h) CJ-CJO alkyl optionally substituted with a substituent selected from the group of hydrogen, ORJO NHR7> NR R9 and SRj \ and (i) C3-Cg cycloalkyl optionally substituted with a substituent selected from the group ofhydrogen, ORιo> NHR7, NRgR9 and SRj \ providing that the compound is not /wyo-inositol. The present invention further provides a method of diagnosing the presence of abnormally folded or aggregated protein and/or amyloid fibril or amyloid in a subject comprising: (a) collecting a sample from said subject; (b) contacting said sample with a radioactive compound or compound tagged with a substance that emits a detectable signal under conditions to allow the binding of said compound to the abnormally folded or aggregated protein and/or amyloid fibril or amyloid if present; and (c) detecting the radioactivity or the signal from the compound bound to the abnormally folded or aggregated protein and/or fibrils or amyloid, thus diagnosing the presence of abnormally folded or aggregated protein and/or amyloid fibril or amyloid in said subject, wherein said compound has the following structure:
Figure imgf000010_0001
wherein each of Rl5 R,.s R2, R2., R3, R3,5 R4s R4,, R5, R5 ,s R6, and R&, is independently selected from the group of;
(a) hydrogen atom;
(b) NHR7, wherein said R7 is selected from the group ofhydrogen; C2- C 10 acyl and C 1 -C \ 0 alkyl;
(c) N gR9, wherein said Rg is C2-C10 acyl or CI -CJO alkyl and said R9 is C2-C10 acyl or C\-C\Q alkyl;
(d) OR] , wherein said R\ is selected from the group of no group, hydrogen, C2-C10 acyl, C I-CJO alkyl and SO3H;
(e) C5-C7 glycosyl;
(f) C3-C cycloalkyl optionally substituted with a substituent selected from the group ofhydrogen, OH, NH2, SH, OSO3H and OPO3H2; (g) SRi i , wherein R\ \ is selected from the group of hydrogen, C \ -C \ Q alkyl and O3H; (h) CJ-CJO au^yl optionally substituted with a substituent selected from the group ofhydrogen, ORJQ, NHR , NR 9 and SRj ; and (i) C3-C cycloalkyl optionally substituted with a substituent selected from the group ofhydrogen, ORιo> NHR7, NRgR9 and SRi 1 providing that the compound is not mvoinositol.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 A shows the structure of myo-, epi- and scyllo-mositol while Figures 1 B- 1 H show the spatial reference memory version of the Morris water maze test in TgCRND8 mice. vø-inositol treatment did not alter cognitive function (IB). At 6 months of age, non-treated TgCRNDS (n=10) show cognitive impairment relative to non-Tg controls and epi- (1C) and scy/Vo-inositol (ID) treated mice (n=10 per group, p<0.02 untreated vs treated). The performance of epz'-inositol treated TgCRNDδ mice remained impaired with respect to non-Tg littermates (IE) whereas the performance of scy/Zo-inositol TgCRNDδ approached that of non-Tg littermates (IF). Non-Tg littermate behavior was not effected by either epi- (1G) or scy/7o-inositol (1H) treatment. Vertical bars represent S.E.M..
Figures 2A-2I show at 6 months of age, the plaque burden and astrogliosis in TgCRNDδ treated with epi- and scy/Zo-inositol treated mice. Control animals have a high plaque load and astrogliosis in the hippocampus (2A) and cerebral cortex (2B). Higher magnification demonstrates that astrocytic activation is not only associated with plaque load (2C). Epz'-inositol treatment has a modest effect on amyloid burden with a decrease in astrogliosis (2D, 2Ε and 2F). Scy/Zo-inositol treatment significantly decreased amyloid burden and gliosis (2G, 2H, and 21). Higher magnification illustrates the smaller mean plaque size in scy/7σ-inositol treated mice (21). Astrocytes were labeled using anti-GFAP antibody and plaque burden was identified using anti- Aβ antibody. Scale Bar 450 microns (A,B,D,E,G,H) and 94 microns (C,F,I).
Figures 3A-3D show that the Aβ species, 1-42, 1-40 and 1-38, in control and treated TgCRND8 mice was indistinguishable (3A) as was the extent of APP processing (3B). Vascular amyloid burden was quantitated on serial sagittal sections in treated and untreated TgCRND8 mice. TgCRNDδ mice have a significant vascular amyloid burden that is associated with small and medium sized vessels, the load is decreased in scy/zO-inositol treated TgCRND8 mice (3A). Scyllo-inositol treatment significantly decreased the total vascular load in comparison to untreated and epi- inositol treated TgCRND8 mice (3C). Sc /Zo-inositol decreases plaque deposition as illustrated by the significant decrease in mean plaque size (3D).
Figure 4 shows the effect of water on the cognitive function of TgCRND8 and non-Tg mice using the spatial reference memory version of the Morris Water Maze in a three day trial paradigm.
Figure 5 shows the effect of scyllo-mosύ.6 on the cognitive function of TgCRND8 and non-Tg mice using the spatial reference memory version of the Morris Water Maze in a three day trial paradigm.
Figure 6 shows the effect of epz'-inositol on the cognitive function of TgCRND8 and non-Tg mice using the spatial reference memory version of the Morris Water Maze in a three day trial paradigm.
Figure 7 shows the effect of w oinositol on the cognitive function of TgCRNDδ and non-Tg mice using the spatial reference memory version of the Morris Water Maze in a three day trial paradigm. Figure 8 shows the effect of scy/fo-inositol, epz'-inositol and røyσ-inositol on the cognitive function of TgCRND8 (learning phase and memory test) and compared with wild type mice using the spatial reference memory version of the Morris Water Maze in a three-day trial paradigm.
Figure 9 shows the percentage of brain area covered with plaques in untreated TgCRND8 mice versus mice treated with scy/Zo-inositol, epz'-inositol or myo-inositol.
Figures 10A and 10B show the survival rates of TgCRNDδ mice treated with water versus epz'-inositol or myo-inositol (10A) or versus •s,cy//ø-inositol (10B).
Figures 11 A-D show the results of spatial reference memory version of the Morris Water Maze test in 6-month old TgCRND8 mice non-treated or treated with mannitol (A,B). Mannitol treated TgCRND8 mice were not significantly different from untreated TgCRNDδ mice (p= 0.89; A). The performance of mannitol treated TgCRNDδ mice was significantly different from mannitol treated non-Tg littermates (p=0.05; B). Plaque burden was analyzed at 6 months of age using quantitative image analyses (C). Mannitol treated TgCRNDδ mice were indistinguishable from untreated TgCRNDδ mice when plaque count was used as a measure of total plaque burden (p=0.87). Vertical bars represent S.E.M.. Kaplan-Meier Cumulative survival plots for TgCRNDδ mice treated and untreated with mannitol (D). The two cohorts of animals, n=35 per group, were not significantly different as determined by the Tarone-Ware statistical test, p=0.87.
Figures 12A and B show the results of a spatial reference memory test in the treatment studies when performed in a 3-day trial paradigm. The performance of scyllo-mositol treated TgCRNDδ mice was comparable to scyllo-inositol treated non- Tg littermates (p=0.38; A). In agreement, scy//o-inositol treated TgCRND8 mice remained indistinguishable from non-Tg littermates after two months of treatment (p=0.67; B).
Figures 13 A and B show Aβ levels within the CNS after administration of various doses of scy//o-inositol were administered once daily for one month to five month old TgCRNDS mice. Soluble Aβ42 levels were decreased at all doses and were significantly different from untreated controls (A). In contrast, insoluble Aβ42 was not significantly different under all conditions (B). Vertical bars represent S.E.M.
Figure 14. TgCRNDδ mice administered various doses of scyllo-inositol once daily for one month were analyzed for levels of brain Aβ40. No difference was detected in soluble (A) and insoluble (B) levels of Aβ40 of untreated and scyllo- inositol treated TgCRNDδ mice at all doses examined.
Figure 15 shows the cognitive performance of 6-month old α//o-inositol- treated TgCRNDδ mice compared with that of their non-transgenic littermates.
Figures 16A-D show that at 2 months of age, the plaque burden in TgPSl x APP mice is decreased in scy/Zo-inositol treated mice. Control animals have a high plaque load in the hippocampus (A) and cerebral cortex (B). Scy/zo-inositol treatment significantly decreased amyloid burden (C, D). Plaque burden identified using anti- Aβ antibody (brown). Scale Bar 300 μm.
Figures 17A-C show the quantification of the plaque burden in TgPSlxAPP mice after -?ςy//o-inositol treatment. The percent brain area covered in plaques (A), mean plaque size (B) and plaque count (C) were significantly reduced. Vertical bars are S.E.M. DETAILED DESCRIPTION OF THE INVENTION
The present invention discloses novel, unpredictable and unexpected properties of certain inositol stereoisomers in relation to the treatment of amyloid- related disorders such as Alzheimer's Disease. It has been surprisingly discovered that certain stereoisomers of inositol and related compounds block Aβ-induced progressive cognitive decline and cerebral amyloid plaque pathology, and improve survival when given to a transgenic mouse model of human Alzheimer Disease during the nascent phase of Aβ deposition.
As disclosed above, previous data suggested that some, but not all, inositol stereoisomers might have an effect on amyloid aggregation in cultured neuronal cells in vitro (McLaurin et al., J. Biol. Chem. 275(24): 18495-18502 (2000)). Those observations did not provide any method to predict which, if any, of the studied stereoisomers (myo-, epi-, scyllo- and c/zz'rø-inositols) would have such effects, nor whether any other stereoisomers would have such effects. Also, those studies could not predict if any inositol stereoisomers would have effects on amyloid deposition, cognitive defects or lifespan in vivo. The present invention describes the unpredictable results that only certain inositol stereoisomers, in particular scyllo- and //σ-inositols reduce amyloid plaque burden, improve cognition and increase lifespan in animal models of amyloid-related disorders, whereas others studied did not have such effects.
Previous studies also suggested only that certain inositol stereoisomers (e.g. epi- and scy//ø-inositols) might inhibit de novo amyloid aggregation in vitro. The present invention describes the unexpected results that scy/Zo-inositol inhibits already established cerebral amyloid deposition, and does so in the living brain. This is not implied by the previously published in vitro data which considered only certain neuronal cell types in culture, not the complex tissues of the living brain, and only suggested that inositols might inhibit de novo aggregation, thereby having no relevance to established disease.
Previous in vitro data also suggested that epi- and scy/7o-inositol administration affects amyloid Aβ40 levels as well as Aβ42 levels. The in vivo dosing study of the present invention revealed the unpredictable result that administration of allo- or scy//o-inositol specifically reduced Aβ42 levels, whereas insoluble Aβ42 and either soluble or insoluble Aβ40 levels were unaffected. The observation of the present invention showing changes in glial activity and inflammation is novel and surprising, and could not have been predicted by the in vitro data previously published.
The observation of the present invention demonstrating that scyZZo-inositol improves lifespan in transgenic model animals is also novel and surprising, since no drug for Alzheimer's Disease has previously been shown to increase survival and extend lifespan in vivo.
Preferably, the compounds of the present invention are 1,2,3,4,5,6- cyclohexanehexols, more preferably selected from the group of cis-, epi-, allo-, muco-, neo- , scyllo-, D-chiro- and L-cAzro-inositols.
Also preferably, these compounds are 1,2,3,4,5-cyclohexanepentols (quercitols), more preferably selected from the group of epi-, vibo-, scyllo-, allo-, talo-, gala-, cis-, muco-, neo-, prøtø-quercitols and enantiomers thereof. Also preferably, these compounds are selected from the group of a cyclohexanetetrol, a cyclohexanetriol, stereoisomer of cyclohexanetetrol, stereoisimer of cyclohexanetriol, enantiomer of cyclohexanetetrol, and enantiomer of cyclohexanetriol.
These compounds may also be compound is pentahydxycyclohexanones or stereoisomers or enantiomers thereof. Yet again preferably, these compounds are inosose compounds selected from the group of scyZZø-inosose, L-c/ϊz'rø-inosose-1 and L-epz'-inosose.
Also preferably, these compounds are trihydroxyxcyclohexanones, or stereoisomers or enantiomers thereof. More preferably, (-)-l- eø y-scyllo-inosose.
Also preferably, these compounds are pentahydxycyclohexanones (inosose), or stereoisomers or enantiomers thereof, more preferably selected from the group of .scy/Zø-inosose, L-c/zz'rø-inosose-l and L-epz'-inosose.
Optionally, these compounds are trihydroxyxcyclohexanones or stereoisomers or enantiomers thereof such as (-)-l-deoxy-scyllo-mosose.
Also preferably, these compounds are O-monomethyl-cyclohexanehexols or stereoisomers or enantiomers thereof, more preferably selected from the group of D- pinitol, L-quebrachitol and D-bornesitol.
Again, these compounds may be selected from the group of , monoaminocyclohexanepentols (inosamines), diaminocyclohexanetetrols (inosadiamines), diaminocyclohexanetriols, stereoisomers thereof, and enantiomers thereof, and pharmaceutically acceptable salts thereof such as L-neø-inosamine, D,L- epz'-inosamine-2, streptamine and deoxystreptamine.
Yet again preferably, these compounds are monomercapto-cyclohexanepentols or stereoisomers or enantiomers thereof, more preferably 1L-1 - eo y-1 -mercapto-8- O-methyl-chiro-inositol.
The most preferred compounds of the present invention are αZZø-inositol and scyZZo-inositol, with scyZZø-inositol being the most preferred. As indicated above, the inositol stereoisomers of the present invention exclude zwyø-inositol and may also exclude epz'-inositol.
Even when given after the amyloid pathology has been well established for several months, these compounds effectively reverse cerebral. Aβ accumulation and amyloid pathology.
Accordingly, these compounds are found to be useful in treating or preventing in a subject a condition of the central or peripheral nervous system or systemic organ associated with a disorder in protein folding or aggregation, or amyloid formation, deposition, accumulation, or persistence. These compounds are also found to be useful in preventing abnormal protein folding, abnormal protein aggregation, amyloid formation, deposition, accumulation, or persistence, or amyloid lipid interactions as well as causing the dissociation of abnormally aggregated proteins and/or dissolving or disrupting pre-formed or pre-deposited amyloid fibril or amyloid in a subject.
Preferably, the condition of the central or peripheral nervous system or systemic organ results in the deposition of proteins, protein fragments and peptides in beta-pleated sheats and/or fibrils and/or aggregates. More preferably, the condition of the central or peripheral nervous system or systemic organ is selected from the group of: Alzheimer's disease, presenile and senile forms; amyloid angiopathy; mild cognitive impairment; Alzheimer's disease-related dementia; tauopathy; α- synucleinopathy; Parkinson's disease; Amyotrophic Lateral Sclerosis; motor neuron Disease; Spastic paraplagia; Huntington's Disease, spinocerebellar ataxia, Freidrich's Ataxia; neurodegenerative diseases associated with intracellular and/or intraneuronal aggregates of proteins with polyglutamine, polyalanine or other repeats arising from pathological expansions of tri- or tetra-nucleotide elements within corresponding genes; cerebro vascular diseases; Down's syndrome; head trauma with post-traumatic accumulation of amyloid beta peptide; Prion related disease; Familial British Dementia; Familial Danish Dementia; Presenile Dementia with Spastic Ataxia; Cerebral Amyloid Angiopathy, British Type; Presenile Dementia With Spastic Ataxia Cerebral Amyloid Angiopathy, Danish Type; Familial encephalopathy with neuroserpin inclusion bodies (FENTJB); Amyloid Polyneuropathy; Inclusion Body myositis due to amyloid beta peptide; Familial and Finnish Type Amyloidosis; Systemic amyloidosis associated with multiple myeloma; Familial Mediterranean Fever; chronic infections and inflammations; and Type II Diabetes Mellitus associate with islet amyloid polypeptide (LAPP). Also preferably, the Alzheimer's disease-related dementias are vascular or
Alzheimer dementia and tauopathy selected from the group of argyrophilic grain dementia, corticobasal degeneration, dementia pugilistica, diffuse neurofibrillary tangles with calcification, frontotemporal dementia with parkinsonism, Prion-related disease, Hallervorden-Spatz disease, myotonic dystrophy, Niemann-Pick disease type C, non-Guamanian Motor Neuron disease with neurofibrillary tangles, Pick's disease, postencephalitic parkinsonism, prion protein cerebral amyloid angiopathy, progressive subcortical gliosis, progressive supranuclear palsy, subacute sclerosing panencephalitis, and tangle only dementia.
Also preferably, the α-synucleinopathy is selected from the group of dementia with Lewy bodies, multiple system atrophy with glial cytoplasmie inclusions, Shy- Drager syndrome, striatonigral degeneration, olivopontocerebellar atrophy, neurodegeneration with brain iron accumulation type I, olfactory dysfunction, and amyotrophic lateral sclerosis.
Again preferably, the Motor Neuron Disease is associated with filaments and aggregates of neuro filament and/or superoxide dismutase proteins, the Spastic paraplegia is associated with defective function of chaperones arid/or triple A proteins and the spinocerebellar ataxia is DRPLA or Machado- Joseph Disease.
Also preferably, the Prion related disease is selected from the group of Creutzfeldt- Jakob disease, Gerstmann-Straussler-Scheinker disease, and variant Creutzfeldt-Jakob disease and the Amyloid Polyneuropathy is Senile amyloid polyneuropathy or Systemic Amyloidosis.
More preferably, the condition of the central or peripheral nervous system or systemic organ is Parkinson's disease including familial and non-familial types. Most preferably, said condition of the central or peripheral nervous system or systemic organ is Alzheimer's disease.
Preferably, the compound is administered to the subject at a dose of about 1 mg to about 1 g per kg, preferably 1 mg to about 200 mg per kg, more preferably about 10 mg to about 100 mg per kg and most preferably about 30 mg to 70 mg per kg of the weight of said subject. The administration can be accomplished by a variety of ways such as orally (oral pill, oral liquid or suspension), intravenously, intramuscularly, intraperitoneally, intraderrnally, transcutaneously, subcutaneously, intranasally, sublingually, by rectal suppository or inhalation, with the oral administration being the most preferred. The administration of the compounds of the present invention can be undertaken at various intervals such as once a day, twice per day, once per week, once a month or continuously.
Preferably, the compounds of the present invention are administered in combination with other treatments such as beta-secretase inhibitors, gamma-secretase inhibitors (APP-specific or non-specific), epsilon-secretase inhibitors (APP-specific or non-specific), other inhibitors of beta-sheet aggregation/fibrillogenesis/ADDL formation (e.g. Alzhemed), NMDA antagonists (e.g. memantine), non-steroidal anti- inflammatory compounds (e.g. Ibuprofen, Celebrex), anti-oxidants (e.g. Vitamin E), hormones (e.g. estrogens), nutrients and food supplements (e.g. Gingko biloba); acetylcholinesterase inhibitors (e.g. donezepil), muscarinic agonists (e.g. AF102B (Cevimeline, EVOXAC), AF150(S), and AF267B), anti-psychotics (e.g. haloperidol, clozapine, olanzapine); anti-depressants including tricyclics and serotonin reuptake inhibitors (e.g. Sertraline and Citalopram Hbr), gene therapy and/or drug based approaches to upregulate neprilysin (an enzyme which degrades Aβ); gene therapy and/or drug based approaches to upregulate insulin degrading enzyme (an enzyme which degrades Aβ), vaccines, immunotherapeutics and antibodies to Aβ (e.g. ELAN AN- 1792), statins and other cholesterol lowering drugs (e.g. Lovastatin and Simvastatin), stem cell and other cell-based therapies, inhibitors of kinases (CDK5, GSK3α, GSK3β) that phosphorylate TAU protein (e.g. Lithium chloride), or inhibitors of kinases that modulate Aβ production (GSK3α, GSK3β, Rho/ROCK kinases) (e.g. lithium Chloride and Ibuprofen).
It is believed that these other therapies act via a different mechanism and may have additive/synergistic effects with the present invention. In addition, many of these other therapies will have mechanism-based and/or other side effects which limit the dose or duration at which they can be administered alone.
Because of their ability to bind amyloids in vivo as discussed hereinbelow in more detail, the compounds of the present invention are also useful in diagnosing the presence of abnormally folded or aggregated protein and or amyloid fibril or amyloid in a subject using a method that comprises administering to said subject a radioactive compound or compound tagged with a substance that emits a detectable signal in a quantity sufficient and under conditions to allow for the binding of said compound to the abnormally folded or aggregated protein and/or fibrils or amyloid, if present; and detecting the radioactivity or the signal from the compound bound to the abnormally folded or aggregated protein and/or fibrils or amyloid, thus diagnosing the presence of abnormally folded or aggregated protein and/or amyloid fibril or amyloid.
Alternatively, a sample suspected of containing abnormally folded or aggregated protein and or amyloid fibril or amyloid is collected from a subject and is contacted with a radioactive compound or compound tagged with a substance that emits a detectable signal under conditions to allow the binding of said compound to the abnormally folded or aggregated protein and/or amyloid fibril or amyloid if present; and thereafter detect the radioactivity or the signal from the compound bound to the abnormally folded or aggregated protein and/or fibrils or amyloid, thus diagnosing the presence of abnormally folded or aggregated. protein and/or amyloid fibril or amyloid in said subject.
Preferably, said detectable signal is a fluorescent or an enzyme-linked iπrmunosorbent assay signal and said sample is whole blood (including all cellular constituents) or plasma. As shown hereinbelow, the compounds of the present invention can abrogate the cerebral accumulation of Aβ, the deposition of cerebral amyloid plaques, and cognitive decline in a transgenic mouse model of Alzheimer Disease when given during the "late presymptomatic" phase, prior to the onset of overt cognitive deficits and amyloid neuropathology in these mice. Furthermore, even when these compounds are given after the onset of cognitive deficits and amyloid plaque neuropathology, they can effectively reverse the amyloid deposition and neuropathology. Importantly, the mechanism of action of these compounds follows a rational design based upon their capacity to modulate the assembly of Aβ monomers into neurotoxic oligomers and/or protofibrils.
Other advantages of the compounds of the present invention include the fact that they are transported into the CNS by both known transporters and by passive diffusion, and therefore provide ready CNS bioavailablility. Second, these compounds are catabolized to glucose. Third, as a class, these compounds generally have low toxicity profiles, and some of them have previously been given to humans albeit for a different purpose.
Example 1 - Development of Alzheimer's mouse model and methods of administering compounds of the present invention TgCRND8 mice are a robust murine model of Alzheimer's disease as described by Janus et al. (Nature 408:979-982 (2000). They express a human amyloid precursor protein (APP695) transgene under the regulation of the Syrian hamster prion promoter on a C3H/B6 outbred background. The human APP695 transgene bears two mutations that cause AD in humans (K670N/M671L and V717F). Beginning at about 3 months of age, TgCRNDδ mice have progressive spatial learning deficits that are accompanied by rising cerebral Aβ levels and by increasing number of cerebral extracellular amyloid plaques that are similar to those seen in the brains of humans with AD (C. Janus et al., Nature 408:979-982 (2000)).
Age and sex-matched cohorts of TgCRNDS mice and non-transgenic littermates (n=35 in each cohort) were either untreated, or were given a compound of the present invention as indicated below at 30mg/day/mouse beginning at age of about 6 weeks. The mice were followed for outcome measures cognitive function, brain Aβ levels, brain pathology, and survival at 4 months and 6 months of age.
Prevention Studies Methods Mice - Experimental groups of TgCRNDδ mice were fed myo-, epi- and sc ZZo-inositol at 30 mg/mouse/day. Two cohorts entered the study at 6 weeks of age and outcomes were analyzed at 4- and 6-months of age. The body weight, coat characteristics and in cage behavior was monitored. All experiments were performed according to the Canadian Council on Animal Care guidelines. Behavioral tests - After non-spatial pre-training, mice underwent place discrimination training for 5 days with 4 trials per day. Behavioral data was analyzed using a mixed model of factorial analysis of variance (ANOVA) with drug or genotype and training sessions as repeated measure factors.
Cerebral amyloid burden - Brains were removed and one hemisphere was fixed in 4% paraformaldehyde and embedded in paraffin wax in the mid saggital plane. To generate sets of systematic uniform random sections, 5μm serial sections were collected across the entire hemisphere. Sets of sections at 50mm intervals were used for analyses (10-14 sections/set). Plaque were identified after antigen retrieval with formic acid, and incubated with primary anti-Aβ antibody (Dako M-0872), followed by secondary antibody (Dako StreptABCcomplex/horseradish kit). End products were visualized with DAB counter-stained with hematoxylin. Amyloid plaque burden was assessed using Leco JA-3001 image analysis software interfaced with Leica microscope and Hitachi KP-M1U CCD video camera. Vascular burden was analyzed similarly and a dissector was used to measure the diameter of affected vessels. Plasma and Cerebral Aβ Content - Hemi-brain samples were homogenized in a buffered sucrose solution, followed by either 0.4% diethylamine/lOOmM NaCI for soluble Aβ levels or cold formic acid for the isolation of total Aβ. After neutralization the samples were diluted and analyzed for Aβ40 and Aβ42 using commercially available kits (BIOSOURCE International). Each hemisphere was analyzed in triplicate with the mean ± SEM reported. Western blot analyses were performed on all fractions using urea gels for Aβ species analyses. Aβ was detected using 6E10 (BIOSOURCE International) and Enhanced Chemiluminenscence (Amersham).
Analysis of APP in brain - Mouse hemi-brain samples were homogenized in 20mM Tris pH7.4, 0.25M sucrose, ImM EDTA and ImM EGTA, and a protease inhibitor cocktail, mixed with 0.4% DEA (diethylamine)/100mM NaCI and spun at 109,000Xg. The supernatants were analysed for APPs levels by Western blotting using mAb 22C11, while the pellets were analysed for APP holoprotein using mAb Cl/6.1. Gliosis Quantitation - Five randomly selected, evenly spaced, sagittal sections were collected from paraformaldehyde-fixed and frozen hemispheres of treated and control mice. Sections were irnrnunolabelled for astrocytes with anti-rat GFAP IgG2a (Dako; diluted 1 :50) and for microglia with anti-rat CD68 IgG2b (Dako; 1 :50). Digital images were captured using a Coolsnap digital camera (Photometries, Tuscon, Arizona) mounted to a Zeiss Axioscope 2 Plus microscope. Images were analysed using Openlab 3.08 imaging software (Improvision, Lexington MA).
Survival Census - The probability of survival was assessed by the Kaplan- Meier technique, computing the probability of survival at every occurrence of death, thus making it suitable for small sample sizes. For the analyses of survival, 35 mice were used for each treatment group. The comparison between treatments was reported using the Tarone-Ware test.
Example 2 - Prevention of cognitive deficits The cognitive function of TgCRNDδ mice was assessed using the spatial reference memory version of the Morris Water Maze using a five-day trial paradigm (Figures 1C-1H). Data from treated and non-treated TgCRNDδ mice, and from treated and non-treated non-Tg littermates (n=10 for all combinations) were analyzed using a mixed model of analysis of variance (ANOVA) with treatment (untreated, epi- or .ycy//ø-inositol) and genotype (TgCRNDδ versus non-Tg) as 'between-subject' factors. TgCRNDδ mice treated with either epi- or _?ςy//ø-inositol performed significantly better than untreated TgCRNDδ mice (p<0.02; Figs. 1C and D). When compared to treated or non-treated non-Tg littermates, epz'-inositol treated TgCRNDδ mice had a slightly slower learning curve during the first three days of training. However, after 4 days of training, epz'-inositol treated TgCRNDS mice were not statistically different from their non-Tg littermates (Fig. 2E). In contrast, scyllo- inositol treated TgCRNDδ mice were indistinguishable from non-Tg littermates on all days. Thus both stereoisomers inhibited the development of cognitive deficits, and scy/Zo-inositol actually prevented the deficits to such a degree that the scyllo-inositol treated TgCRND8 mice were indistinguishable from normal mice. This improved performance was not due to a non-specific effect on behavioral, motoric, or perceptual systems because epi- and scy//ø-inositol treatment had no effect on the performance of non-Tg mice (Figures 2G and 2H). The improved performance was also not due to nutritional or caloric effects because body weight, activity, and coat condition were not different between treated and untreated cohorts. Furthermore, treatment with mannitol (a sugar of similar molecular weight) had no effect on behavior. Gender effects were not significant between any treatment group (p=0.85). Example 3- Reduction of cerebral Aβ Burden and Amyloid Neuropathology
At four months of age, untreated TgCRND8 mice have a robust expression of both Aβ40 and Aβ42 (Table 1). Epz'-inositol treatment as described in Example 1 reduced both Aβ 40 (43±2% reduction in both soluble and insoluble pools; p<0.05) and Aβ 42 levels (69% reduction in soluble pool, p=0.005; 28% reduction in insoluble pool, p=0.02) at 4-months of age. However, these improvements were not sustained, and by 6 months of age, brain Aβ levels rose to levels similar to those observed in untreated TgCRND8 mice (Table 1).
In contrast, at four months of age, scyZZoinositol treatment decreased total brain Aβ40 by 62% (p=0.0002) and total brain Aβ42 by 22% (p=0.0096; Table 1). At 6 months of age, ^cyZZø-inositol treatment caused a 32% reduction in Aβ40 levels (ρ=0.04) and 20% reduction in Aβ42 (ρ=0.02) compared to untreated TgCRND8 mice.
Because the decreased Aβ concentrations detected after inositol treatment could have resulted from altered efflux of Aβ into the plasma, Aβ-β levels in the plasma were examined at 4- and 6-months of age (Table 1). TgCRNDδ mice have high plasma Aβ concentrations at 4-months of age and remain constant at 6 months of age even though CNS plaque load is still rising at 6-months of age (Table 1). Neither epz'-inositol nor sey/Zo-inositol treatment had any effect on plasma Aβ levels in comparison to untreated TgCRNDS mice (p-0.89). The most parsimonious explanation for this observation is that the inositols have selectively altered the fibrillization of Aβ in the CNS, but have not affected β- or γ-secretase activity, or the normal mechanisms for clearance of Aβ into plasma. Nevertheless, this observation is significant for two reasons. First, a drop in plasma and CSF Aβ levels is usually detected as the clinical course progresses in untreated AD patients (Mayeux, et al, Ann. Neurol 46, 412, 2001). Secondly, patients in the AN1792 immunization study who developed a strong antibody response and an apparent clinical response did not have altered plasma Aβ-β levels (Hock et al, Neuron 38, 547 2003). Therefore, these results indicate that it is not necessary to change plasma Aβ levels to obtain an effective therapeutic outcome.
To confirm that inositol stereoisomers had no effect on either the expression or proteolytic processing of APP, the levels of APP holo-protein, sAPP-α, and various Aβ species were examined within the brain of inositol-treated and untreated TgCRND8 mice. Consistent with our previously reported data (McLaurin, et al, Nat. Med. 8, 1263, 2002), Aβ42, Aβ40 and Aβ38 are the predominant species in the brain of TgCRNDδ mice (Figure 3 A), and the CNS levels of immature and mature glycolyslated APP (Figure 3B), and of sAPP-α were indistinguishable regardless of treatment. In combination, these results indicate that epi- and scyllo-inositol have a direct and selective effect on Aβ oligomerization and not the processing of APP.
The changes in Aβ-β peptide load were accompanied by a significant decrease in plaque burden (Table 1; Figures 2A-2I). In epi-inositol treated TgCRNDδ mice, there was a significant decrease in the mean plaque size at 4- but not 6-months of age compared with untreated TgCRNDδ mice (95 ± 4.3 μm2 versus 136 ± 15μm2, p = 0.04; 370 ± 9μm2 versus 423 ± 22μm2, p = 0.06, respectively). These results indicate that at modest Aβ levels, epz'-inositol prevents Aβ oligomerization but once initiated at higher Aβ concentrations, epz'-inositol is unable to inhibit fibrillogenesis. Scyllo- inositol treatment decreased the mean plaque size from 136±15 μm2 to 103±4 μm2 (p=0.01) at 4 months of age. In .scyZZo-inositol treated TgCRNDδ mice at 6 months of age, the decrease in Aβ peptide levels was accompanied by a 20% reduction in plaque number (p = 0.005), a 35% decrease in brain area covered with plaques (p = 0.015) and a decreased mean plaque size (339 ± 10 vs. 423 ± 21μm2, p = 0.009). These results demonstrate that by every measure there was a reduction in plaque burden after scyllo-inositol treatment.
Table 1. Inositol treatment decreases Aβ40 and Aβ42 Levels
Aβ40 Aβ42 Plaque Total Plaque
(ng/gm wet brain ± sem) (ng/gm wet brain ± sem) Total Plaque Area Area/Total
Soluble Insoluble Soluble Insoluble Aβ Count (μm2) Brain Area
CΛ (%)
C 00 CΛ 4 month prevention
Control 75±6 1163±9 273±18 5658±248 7169±284 696±25 100766±7564 0.026±0.004
Epi-Inositol 43±7* 615±32t 85±7| 4059±179* 4802±176 678±64 65042±5199 0.020±0.001 m
CΛ Scyllo-inositol 37±5* 437±80| 206±8* 4409±135* 5089±173 598±19* 63847±2895 0.015±0.001*
I m 6 month prevention m
Control 187±29 3576±172 626±87 15802±237 20191±211 960±44 411288±11912 0.120±0.001
73 Epi-Inositol 188±24 3668±149 665±39 13943±277f 18464±229 979±32 380456*13498 0.096±0.04 m Scyllo-inositol 105±8* 2453±251*f 475±26* 12588±82f 15621±151 774±10*f 262379±5373f 0.079±0.013t r σ> Plasma Aβ Levels
(pg/ml)
4 month prevention 6 month prevention
Control 1018±27 915±59
Epi-Inositol 1082±164 952±56
Scyllo-inositol 952±49 905±55
Anova with Fisher's PLSD, f p<0.001 and * p<0.05
Example 4 - Reduction of glial reactivity and inflammation
Astroglial and microglial reactions are neuropathological features both of human AD and of all amyloid mouse models (Irizarry et al., J Neuropathol Exp Neurol 56 , 965, 1997; K. D. Bornemann et al. Ann N Y Acad Sci. 90δ, 260, 2000). Therefore, the effect of epi- and scy/Zø-inositol treatment was investigated on astrogliosis and microgliosis in the brains of TgCRND8 mice (Figures 3A-3D). Serial sagittal sections were stained with the astrocytic marker glial fibrillary acidic protein (GFAP) and quantitated for percent brain area covered by astrogliosis. TgCRNDδ mice have a high basal astrogliosis at 4-months of age (0.459±0.04δ%), which increases slightly by 6-months of age (0.5δ4±0.0δ9%), and which is not restricted to plaque areas (Figures 2A-C). Epz'-inositol decreased the astrogliotic response to 0.388±0.039% at 6-months of age (p=0.04; Fig. 2D-F). Scyllo- inositol, on the other hand, decreased astrogliosis much more efficiently to 0.269±0.02δ% at 6-months of age, (p=0.006)(Fig. 2G-I). Microglial activation was also significantly attenuated in scy//σ-inositol treated TgCRNDδ mice (0.20± 0.008% brain area) when compared to age- and sex-matched untreated TgCRNDS mice (0.31 ± 0.01%; pθ.001). However, epz'-inositol treated mice demonstrated no significant reduction in microglial activation at 6 months (0.24δ ± 0.02%; p= NS). Taken together these data indicate that scyZ/o-inositol treatment decreases the Aβ -induced inflammatory response within the CNS.
Example 5 - Reduction of vascular amyloid load
Alzheimer's disease is characterized by the presence of both parenchymal and vascular amyloid deposits. In untreated 6 month old TgCRNDδ mice approximately 0.03% of the brain area is associated with vascular amyloid. No difference could be detected in the vascular amyloid burden after epz'-inositol treatment at 6 months of age (Figure 3C). In contrast, scy/Zo-inositol treatment significantly decreased the vascular amyloid burden (p=0.05) (Fig. 3C), and the amyloid deposition was predominantly localized to smaller vessels, < 25 m2 in diameter (56 ± 2% versus 70 ± δ% in small vessels in untreated TgCRNDδ mice). The mean size of cerebrovascular plaques was significantly decreased in the scyllo-inositol treated mice in comparison to untreated mice (154±16 vs. 363±34, p=0.008; Figure 3D).
Example 6 - Survival improvement
TgCRNDδ mice have a 50% survival at 175 days, which after treatment was improved to 72% with scy/Zø-inositol (n=35 per group, p<0.02 for scvZZø-inositol vs. control, Figure 10B). Treatment with ø-inositol did not affect overall survival significantly (Figure 10A). Control experiments confirmed that the enhanced survival of cyZZo-inositol treated mice was not an indirect effect of increased caloric intake. Thus, treatment of wild type mice with sc ZZo-inositol had no effect either on survival or on other parameters such as weight, fur condition or cage behavior. Furthermore, the weight, fur condition and home-cage behavior of the inositol-treated TgCRNDδ mice did not vary from untreated TgCRNDδ mice. Simultaneous experiments with mannitol, a simple sugar of similar molecular weight, also had no effect on survival of TgCRNDδ mice.
Example 7 - Treatment and Reversal of amyloid deposition Taken together, the prevention studies demonstrate that scyllo-inositol inhibits both parenchymal and cerebrovascular amyloid deposition and results in improved survival and cognitive function in the TgCRND8 mouse model of Alzheimer disease. However, most Alzheimer's disease patients will likely seek treatment only once symptomatic, and when Aβ oligomerization, deposition, toxicity and plaque formation are already well advanced within the CNS. A pilot study was therefore initiated on 5 month old TgCRNDδ mice. These mice have significant Aβ and plaque burdens that are comparable to those in the brain of humans with AD.
Treatment Study Methods
Mice - Experimental groups of TgCRNDδ mice were fed myo-, epi- and scyllo- inositol at 30 mg/mouse/day. A cohort entered the study at 5 months of age and outcomes were analyzed at 6-months of age. The body weight, coat characteristics and in cage behavior was monitored. All experiments were performed according to the Canadian Council on Animal Care guidelines.
Survival Census - The probability of survival was assessed by the Kaplan-Meier technique, computing the probability of survival at every occurrence of death, thus making it suitable for small sample sizes. For the analyses of survival, 35 mice were used for each treatment group. The comparison between treatments was reported using the Tarone-Ware test.
Behavioral Test - Reversal Study - Mice entered the Morris water maze test with a hidden platform on day one without pretraining. Mice were tested for 3 days with six trials per day. On the fourth day, the platform was removed from the pool and each mouse received one 30-s swim probe trial. On the last day the animals underwent a cue test in order to evaluate swimming ability, eye sight and general cognition. The cue test is composed at the platform being placed in a different quadrant than that used for testing and is tagged with a flag. Animals are allowed 60 s to find the platform. Animals that do not find the platform are not used in the final analyses of spatial memory. Behavioural data was analysed using a mixed model of factorial analysis of variance (ANOVA) with drug or genotype and training sessions as repeated measure factors.
Cerebral amyloid burden - Brains were removed and one hemisphere was fixed in 4% paraformaldehyde and embedded in paraffin wax in the mid saggital plane. To generate sets of systematic uniform random sections, 5μm serial sections were collected across the entire hemisphere. Sets of sections at 50mm intervals were used for analyses (10-14 sections/set). Plaque were identified after antigen retrieval with formic acid, and incubated with primary anti-Aβ antibody (Dako M-0872), followed by secondary antibody (Dako StreptABCcomplex/horseradish kit). End products were visualized with DAB counter-stained with hematoxylin. Amyloid plaque burden was assessed using Leco IA-3001 image analysis software interfaced with Leica microscope and Hitachi KP-MIU CCD video camera.
Plasma and Cerebral Aβ Content - Hemi-brain samples were homogenized in a buffered sucrose solution, followed by either 0.4% diethylamine/lOOmM NaCI for soluble Aβ levels or cold formic acid for the isolation of total Aβ. After neutralization the samples were diluted and analyzed for Aβ40 and Aβ42 using commercially available kits (BIOSOURCE International). Each hemisphere was analyzed in triplicate with the mean ± SEM reported.
Results and Significance - All animals that entered the reversal study survived and did not display outward signs of distress or toxicity. The cognitive function of TgCRNDS mice was assessed using the spatial reference memory version of the Morris Water Maze using a three day trial paradigm (Figures 4-δ). Data from treated and non- treated TgCRNDδ mice, and from treated and non-treated non-Tg littermates (n=10 for all combinations) were analyzed using a mixed model of analysis of variance (ANOVA) with treatment (untreated, myo-, epi- or scyllo-inositol) and genotype (TgCRND8 versus non-Tg) as 'between-subject' factors. In this paradigm TgCRND8 mice were significantly impaired in comparison to wild type littermates (Figure 4). In contrast, ,sey//o-inositol treated TgCRNDδ mice were indistinguishable from non-Tg littermates on all days. (p=0.38; Figure 5). When compared to treated non-Tg littermates, epz'-inositol treated TgCRND8 mice were almost significantly different (p=0.07; Figure 6). Similarly, myo-inositol treated TgCRNDδ mice were significantly different from treated non-Tg littermates (p=0.05, Figure 7). When the learning phase of the Morris water maze test is compared between treatments, all mice behaved similarly (Figure 8). In contrast, only scvZZo-inositol was indistinguishable from non-Tg littermates (Figure 8). Thus, scyllo- inositol actually reversed the cognitive deficits to such a degree that the sc Z/σ-inositol treated TgCRND8 mice were indistinguishable from normal mice. This improved performance was not due to a non-specific effect on behavioral, motoric, or perceptual systems because epi- and scyllo-inositol treatment had no effect on the performance of non-Tg mice. The improved performance was also not due to nutritional or caloric effects because body weight, activity, and coat condition were not different between treated and untreated cohorts.
In order to determine if the improved cognition was associated with decreased plaque burden and Aβ load, brain tissue was examined post-mortem. The changes in cognition were accompanied by a corresponding change in plaque burden and Aβ load (Figure 9 and Table 2). JWyσ-inositol treatment did not affect the plaque burden or Aβ load (Figure 9 and Table 2). In epz'-inositol treated TgCRNDδ mice, there was not a significant decrease in the mean plaque size compared with untreated TgCRNDδ mice (Figure 9), yet the Aβ load was significantly decreased (Table 2). These results suggest that at modest Aβ levels, epz'-inositol prevents Aβ oligomerization but at higher Aβ concentrations, epz'-inositol is unable to inhibit fibrillogenesis completely. Scyllo-inositol treatment significantly decreased the plaque burden and the Aβ load. These results demonstrate that by every measure there was a reduction in plaque burden after scyllo- inositol treatment. These results are comparable in effect size to the 6-month prophylactic studies, and further support the potential for scyllo-inositol.
Because the decreased Aβ concentrations detected after inositol treatment could have resulted from altered efflux of Aβ into the plasma, we examined Aβ levels in the plasma (Table 2). TgCRND8 mice have high plasma Aβ concentrations at 6 months of age. Neither myo-inositol, epi-inositol nor scyllo-inositol treatment had any effect on plasma Aβ levels in comparison to untreated TgCRND8 mice (p=0.89). The most parsimonious explanation for this observation is that the inositols have selectively altered the fibrillization of Aβ in the CNS, but have not affected β- or γ-secretase activity, or the normal mechanisms for clearance of Aβ into plasma. Nevertheless, this observation is significant for two reasons. First, a drop in plasma and CSF Aβ levels is usually detected as the clinical course progresses in untreated AD patients. Secondly, patients in the AN2792 immunization study who developed a strong antibody response and an apparent clinical response did not have altered plasma Aβ levels. Therefore, these results further indicate that it is not necessary to change plasma Aβ levels to obtain an effective therapeutic outcome.
Taken together, these data reveal that selected sey/Zσ-inositol can abrogate the cerebral accumulation of Aβ, the deposition of cerebral amyloid plaques, and cognitive decline in a transgenic mouse model of Alzheimer Disease when given during the "late presymptomatic" phase, prior to the onset of overt cognitive deficits and amyloid neuropathology in these mice. Furthermore, even when scyZZo-inositol is given after the onset of cognitive deficits and amyloid plaque neuropathology, these compounds can effectively reverse the amyloid deposition, neuropathology and cognitive deficits. Therefore, these results indicate that scyZZo-inositol is effective at both prevention of disease and in the treatment of existing disease in patients already diagnosed with AD. Table 2. Inositol treatment decreases Aβ40 and Aβ42 Levels
Aβ40 Aβ42 Plaque Total Plaque
(ng/gm ^ wet brain ± sem) (ng/gm wet brain ± sem) Total Plaque Area Area/Total Brain Aβ Count (μm2) Area (%) Soluble Insoluble Soluble Insoluble
4 month prevention
Control 75±6 1163±9 273±18 5658±248 7169±284 696*25 100766*7564 0.026±0.004
CΛ Myo-Inositol 42±6 485±143 174±9 4268±308 4969±434 649±50 91902*7453 0.023±0.004
C Epi-Inositol 00 43±7* 615±32f 85±7f 4059±179* 4802±176 678±64 65042*5199 0.020*0.001 CΛ Scyllo-inositol 37±5* 437±80f 206±8* 4409±135* 5089±173 598*19* 63847*2895 0.015*0.001*
6 month prevention m Control 187±29 3576±172 626±87 15802±237 20191±211 960±44 411288*11912 0.120±0.001
CΛ Myo-Inositol 221±19 3436±189 543±71 13289±535 17489*354 927±78 400013*19638 0.100*0.005
I Epi-Inositol 188±24 3668±149 665±39 13943±277f 18464*229 979±32 380456*13498 0.096±0.04 m m Scyllo-inositol 105±8* 2453±251*f 475±26* 12588±82t , 15621*151 774±10*| 262379±5373f 0.079±0.013f
1 month treatment
73 c Control 207±16 4965±457 426±14 14503*1071 20101±854 1441*29 486002*16156 0.159±0.014 m Myo-Inositol 194±12 4187±226 487±25 15622±675 20490±526 1324±69 469968*35664 0.153*0.088 r Epi-Inositol 264±11 3637±113 540±14 12830±330 17271*415 1342*114 459706*49966 0.134±0.017 σ> Scyllo-inositol 178±11 3527±241 374±23 11115±647 15194*579 1260±27* 420027*14986* 0.119±0.010*
Plasma Aβ Levels (pg/ml)
4 month prevention 6 month prevention 1 month treatment
Control 1018±27 915±59 2287*151 Myo-Inositol 942±30 969±67 2110*174 Epi-Inositol 1082±164 952±56 2158±157 Scyllo-inositol 952±49 905±55 1980*146
Anova with Fisher's PLSD, f p<0.001 and * ρ<0.05; IP=in progress.
Example 8 - Two-month treatment study with Scy/to-inositol
In order to determine longer efficacy ranges of -scy/Zo-inositol for the treatment of disease, 5-month old TgCRNDδ mice were fed scyllo-inositol or untreated for two months (n=10 per group). The cognitive function of 7-month old TgCRNDδ mice treated with scyZZo-inositol was compared to untreated TgCRNDδ and treated non-Tg littermates in the three-day paradigm of the Morris Water Maze. Behavioural data was analysed using a mixed model of factorial analysis of variance (ANOVA) with drug and genotype as between subject variables and training sessions as within subject variable. As was seen with the 1 -month treatment of scy/Zσ-inositol (Fig. 12A), TgCRNDδ mice treated for two months with scyllo-inositol were indistinguishable from scyZ/o-inositol treated non-Tg littermates (Fig. 12B). In order to correlate the improved cognition with pathology, Aβ40 and Aβ42 levels were analysed in the brain (Table 3). Both insoluble Aβ40 and Aβ42 levels were decreased 20% after scyllo-inositol treatment. These results demonstrate that scyllo-inositol effects persist during disease progression.
Table 3. Inositol treatment decreases Aβ40 and Aβ42 Levels
Brain Aβ40 Brain Aβ42 Plasma Aβ Levels (ng/gm wet brain ± sem) (ng/gm wet brain ± sem) (pg/ml)
Soluble Insoluble Soluble Insoluble Aβ40 Aβ42
2 month treatment
Control 487*14 6924*287 764*51 25827*1238 5212*219 3455*331 Scyllo-mositol 395*60 5703*612* 688*28 20818*1404* 4507*207 3035*236
ANOVA with Fisher's PLSD, * p<0.05.
Example 9 - Effect of Dose on Pathological Outcome in Disease Bearing TgCRND8 mice.
5-month old TgCRNDδ mice were gavaged once daily with scyZ7o-inositol in water at doses of 10 mg/Kg, 30 mg/Kg, 100 mg/Kg or untreated. Animals were sacrificed after one month of treatment and analysed for pathological outcomes. Analysis of the levels of Aβ within the brain of all the cohorts demonstrates that all drug doses were effective to the same extent on lowering soluble Aβ42 levels in comparison to untreated TgCRNDδ mice (20% reduction, F3>15=3.1, p=0.07; Fig. 13A). Analyses of individual doses demonstrate that 10 mg/Kg and 30 mg/Kg doses were significantly different from untreated controls (p=0.03 and p=0.02, respectively). None of the doses chosen were significantly different from each other (F2 ,,=0.6, p=0.57; Fig. 13 A). Gavage dosing had no significant effect on insoluble Aβ42 (F3 15=0.69, p=0.5δ; Fig. 13B) or soluble and insoluble Aβ40 (F3>]5=0.04, p=0.99 and F3>15=0.36, ρ=0.79, respectively; Fig. 14A andl 4B).
Example 10 - Efficacy of α/fo-inositol for the treatment of disease bearing TgCRND8 mice.
To assess whether αZZo-inositol might also be effective in preventing further progression and/or might partially reverse a well-established AD-like phenotype, the start of treatment of the TgCRNDδ mice was delayed until 5 months of age. Cohorts of TgCRNDδ and non-transgenic littermates were either treated for 2 days with allo- inositol, or were untreated. In these experiments, the dosage and oral administration of compounds, and the behavioral and neurochemical assays were the same as those employed in the above treatment experiments.
The cohort of 6-month old αZ/ø-inositol-treated TgCRNDδ mice performed significantly better than untreated TgCRNDδ mice (F, ]3=0.45, p=O.05; data not shown). The cognitive performance of 6-month old allo-inositol-treated TgCRNDδ mice was still significantly different from that of their non-transgenic littermates (FU3=5.9, p=0.05; Fig. 15). The beneficial effect of inositol treatment was not due to non-specific effects on behavioral, motor, or perceptual systems because inositol treatment had no effect on the cognitive performance of non-Tg mice (F, ,2=0.9δ; p=0.49). Cerebral Aβ levels were analyzed for treatment versus untreated TgCRNDδ mice to determine whether improved behavior could be correlated with changes in Aβ (Table 4). ^ZZo-inositol treatment reduced soluble Aβ42 (20% reduction, p<0.05) an effect similar to that seen for scyllo- inositol. yϊZZø-inositol did not significantly alter insoluble Aβ42 or Aβ40 (soluble and insoluble pools). One possible explanation for the decrease in Aβ42 is clearance of Aβ42 in the periphery with a subsequent increase in plasma Aβ42. The levels of Aβ42 in plasma after α/Zo-inositol treatment were indistinguishable from untreated TgCRNDδ plasma levels (Table 5). In agreement with the other inositol stereoisomers, these results demonstrate that plasma Aβ levels are unaffected by α/Zo-inositol treatment.
Table 4. Allo-Inositol treatment decreases Aβ42 levels
Brain Aβ40 Brain Aβ42 Plasma Aβ Levels (ng/gm wet brain ± sem) (ng/gm wet brain ± sem) (pg/ml)
Soluble Insoluble Soluble Insoluble
1 month treatment
Control 252*48 4105*851 666*39 16448*2120 2359*147
Allo-inositol 281*21 3787*342 547*47* 16336*910 2458*95
ANOVA with Fisher's PLSD, * p<0.05.
Table 5. Blood Biochemistry - scyZ/ø-inositol Dose Study
Untreated 100 mg/Kg 30 mg/Kg 10 mg/Kg Reference Levels (Vita-Tech & CCAC) n=4 n=4 n=3 n=5 iochemistry Total protein 46*2 g/L 49*2 50*2.6 50*3 35-72
Albumin 35*0 g/L 31*1 33*2 33*4 25-48
Globulin 12*1 g/L 19*2 17*1 17*2 18-82
Bilirubin 2.4±lumol/L 1.9*0 2.0*1 1.9*0.6 2-15
ALP 81*10 U/L 76*11 81*10 73*22 28-94
ALT 42*4 U/L 38*4 42*4 51*20 28-184
Glucose ll±2 mmol/L 11*2 12*2 7*2 9.7-18.6
Urea 9*3 mmol/L 7.4*1 9*3 10*2 12.1-20.6
Creatinine 36*5 umol/L 31*4 35*5 40*5 26-88
Hemolysis Normal Normal Normal Normal
Icteria Normal Normal Normal Normal
Lipemia Normal Normal Normal Normal
E∑ample 11 - Inositol Treatment does not affect Blood Chemistry
In order to rule out any deleterious effects of inositol treatment on blood chemistry and organ function, blood was analyzed after one month treatment with both scyllo- and allo-inositol (Table 5,6). The total protein, albumin, globulin, bilirubin, alkaline phosphatase, glucose, urea and creatinine were not significantly different between treatment groups or from untreated TgCRNDδ mice. All levels fell within the normal range as determined for non-transgenic wild type mice. In addition hemolysis, icteria and lipemia were all normal. These results suggest that allo- and
Figure imgf000035_0001
do not exhibit obvious deleterious effects on blood chemistry or organ function. Table 6. Blood Biochemistry - 1 Month Treatment Study
Untreated Allo-Inositol Reference Levels (Vita-Tech & CCAC) n=4 n=4
Biochemistry
Total protein 46*2 g/L 48*2 35-72
Albumin 35*0 g/L 32*2 25-48
Globulin 12*1 g/L 17*3 18-82
Bilirubin 2.4±lumol/L 2.9*3 2-15
ALP 81*10 U/L 95*16 28-94
ALT 42*4 U/L 44*4 28-184
Glucose 11*2 mmol/L 10*3 9.7-18.6
Urea 9*3 mmol/L 18.6*13 12.1-20.6
Creatinine 36*5 umol L 69*64 26-88
Hemolysis Normal Normal
Icteria Normal Normal
Lipemia Normal Normal
Example 12 - Efficacy of sci'llø-mmitnl In preventing A -like pathology in a double transgenic mouse model of Ateheimer'g disease., PSl ∑ APP
Tg PSl x APP mice are an enhanced model of Alzheimer's disease which express a mutant human PSl transgene encoding two familial mutations (M146L and L2δ6V) in conjunction with the human APP transgene encoding the Indiana and Swedish familial mutations. These animals develop robust expression of cerebral Aβ levels and amyloid deposition by 30-45 days of age. In a prophylactic trial, TgPSlxAPP mice were treated with -fcy/Zø-inositol from weaning and were assessed for effects on neuropathology at 2 months of age (Figures 16 and 17). Compared with untreated TgPSlxAPP mice, scyllo- inositol treated TgPSlxAPP mice displayed a significant decrease in all measures of plaque burden at 2 months of age. (% brain area covered in plaques= 0.157±0.007 vs 0.065±0.016, pO.OOl; mean plaque size = 177±δ μm2 vs 149±5 μm2, p<0.05; plaque count 3054±324 vs 1514±510, ρ<0.01; (Fig. 17). These results demonstrate that scyllo- inositol prevents amyloid deposition in two robust models of Alzheimer's disease.
Example 13 - Effect of increased caloric intake on TgCRND8 mice In order to rule out the contribution of increased caloric intake or non-specific effects, TgCRNDδ mice were treated with a simple sugar of similar molecular weight, mannitol. At 6 months of age, mannitol treated TgCRNDδ mice were indistinguishable from untreated TgCRNDδ mice (Fig. 11 A) and were significantly different from mannitol treated non-Tg littermates (Fig. 1 IB). Mannitol had no effect on the behaviour of non-Tg mice, since mannitol treated non-Tg mice were indistinguishable from untreated non-Tg mice. These results correlate with the pathological studies that indicate mannitol did not alter the plaque load in TgCRNDδ mice (Fig. 11 C). Simultaneous monitoring of survival demonstrated that mannitol had no effect on the survival of TgCRNDδ mice (Fig. 1 ID). Although the present invention has been described in relation to particular embodiments thereof, many other variations and modifications and other uses will become apparent to those skilled in the art. The present invention therefore is not limited by the specific disclosure herein, but only by the appended claims.

Claims

WHAT IS CLAIMED IS:
1. A method of treating or preventing in a subject a condition of the central or peripheral nervous system or systemic organ associated with a disorder in protein folding or aggregation, or amyloid formation, deposition, accumulation, or persistence comprising administering to said subject a pharmaceutically effective amount of a compound having the following structure:
Figure imgf000038_0001
wherein each of R„ Rr, R2, R2,, R3, R3., R4, R4., R5, R5 ,, R6, and R6> is independently selected from the group of: . (a) Irydrogen atom; (b) NHRy, wherein said R is selected from the group ofhydrogen; C2-C10 acyl and
Figure imgf000038_0002
aϊkylj
(c) NRgRo, wherein said Rg is C2-C10 acyl or Cχ-C Q alkyl and said R9 is C2-C10 acyl or C1-C10 alkyl;
(d) ORιo> wherein said Rjrj is selected from the group of no group, hydrogen, C2-C10 acyl, C ] -Cj Q alkyl and SO3H;
(e) C5-C7 glycosyl;
(f) C3-Cg cycloalkyl optionally substituted with a substituent selected from the group ofhydrogen, OH, NH2, SH, OSO3H and OPO3H2;
(g) SRi 1, wherein R\ is selected from the group ofhydrogen, Cj-Ci Q alkyl and O3H;
(h) Cj-Cio alkyl optionally substituted with a substituent selected from the group of hydrogen, ORJO* NHR7> NRgRo. and SRj \; and (i) C3~C cycloalkyl optionally substituted with a substituent selected from the group ofhydrogen, ORJQ, NHR7, NRgRo, and SRi \ providing that the compound is not myo-inositol.
2. The method of claim 1, wherein said compound is said compound is a 1 ,2,3,4,5,6-cyclohexanehexol.
3. The method of claim 2, wherein said compounds is selected from the group of cis-, epi-, allo-, muco-, neo-, scyllo-, Ω-chiro- and L-c/jz'ro-inositols.
4. The method of claim 1, wherein said compound is a 1,2,3,4,5- cyclohexanepentol .
5. The method of claim 4, wherein said compound is a quercitol selected from the group of epi-, vibo-, scyllo-, allo-, talo-, gala-, cis-, muco-, neo-, proto- quercitols and enantiomers thereof.
6. The method of claim 1 , wherein said compound is selected from the group of a cyclohexanetetrol, a cyclohexanetriol, stereoisomer of cyclohexanetetrol, stereoisimer of cyclohexanetriol, enantiomer of cyclohexanetetrol, and enantiomer of cyclohexanetriol.
7. The method of claim 1, wherein said compound is a pentahydroxycyclohexanone or a stereoisomer or an enantoimer thereof. δ. The method of claim 7, wherein said compound is an inosose selected from the group of scyllo-inosose, L-c/zz>ø-inosose-l and L-e/w'-inosose.
9. The method of claim 1, wherein said compound is a trihydroxycyclohexanone, or a stereoisomer or an enantiomer thereof.
10. The method of claim 9, wherein said compound is (-)-\-deoxy-scyllo- inosose.
11. The method of claim 1 , wherein said compound is a pentahydroxycyclohexanone, or a stereoisomer or an enantiomer thereof.
12. The method of claim 11 , wherein said compound is an inosose selected from the group of _?ςy//e>-inosose, L-c 'ro-inosose-1 and L-ep?'-inosose.
13. The method of claim 1 , wherein said compound is a trihydroxyxcyclohexanone or a stereoisomer or an enantiomer thereof.
1 . The method of claim 13, wherein said compound is (-)-l -deoxy-scyllo- inosose.
15. The method of claim 1, wherein said compound is an O-monomethyl- cyclohexanehexol or a stereoisomer or an enantiomer thereof.
16. The method of claim 15, wherein said compound is selected from the group of D-pinitol, L-quebrachitol and D-bornesitol.
17. The method of claim 1 , wherein said compound is selected from the group of monoaminocyclohexanepentols (inosamines), diaminocyclohexanetetrols (inosadia ines), diaminocyclohexanetriols, stereoisomers thereof and enantiomers thereof, and pharmaceutically acceptable salts thereof.
1 δ . The method of claim 17, wherein said compound is selected from the group of L-πeo-inosamine, DL-epf-inosamine-2, streptamine and deoxystreptamine.
19. The method of claim 1 , wherein said compound is a monomercapto- cyclohexanepentol or a stereoisomer or an enantiomer thereof
20. The method of claim 19, wherein said compound is L- 1 -deoxy- 1 - mercapto-8-O-methyl-chiro-inositol.
21. The method of claim 1 , wherein said compound is scyllo-mositol.
22. The method of claim 1 , wherein said compound is αZ/o-inositol.
23. The method of claim 1 , wherein said condition of the central or peripheral nervous system or systemic organ results in the deposition of proteins, protein fragments and peptides in beta-pleated sheats and/or fibrils and/or aggregates.
24. The method of claim 23, wherein said condition of the central or peripheral nervous system or systemic organ is selected from the group of: Alzheimer's disease, presenile and senile forms; amyloid angiopathy; mild cognitive impairment; Alzheimer's disease-related dementia; tauopathy; α-synucleinopathy; Parkinson's disease; Amyotrophic Lateral Sclerosis; motor neuron Disease; Spastic paraplegia; Huntington's Disease, spinocerebellar ataxia, Freidrich's Ataxia; neurodegenerative diseases associated with intracellular and/or intraneuronal aggregates of proteins with polyglutamine, polyalanine or other repeats arising from pathological expansions of tri- or tetra-nucleotide elements within corresponding genes; cerebrovascular diseases; Down's syndrome; head trauma with post-traumatic accumulation of amyloid beta peptide; Prion related disease; Familial British Dementia; Familial Danish Dementia; Presenile Dementia with Spastic Ataxia; Cerebral Amyloid Angiopathy, British Type; Presenile Dementia With Spastic Ataxia Cerebral Amyloid Angiopathy, Danish Type; Familial encephalopathy with neuroserpin inclusion bodies (FENTB); Amyloid Polyneuropathy; Inclusion Body myositis due to amyloid beta peptide; Familial and Finnish Type Amyloidosis; Systemic amyloidosis associated with multiple myeloma; Familial Mediterranean Fever; chronic infections and inflammations; and Type II Diabetes Mellitus associate with islet amyloid polypeptide (LAPP).
25. The method of claim 24, wherein said condition of the central or peripheral nervous system or systemic organ is Alzheimer's disease.
26. The method of claim 24, wherein said Alzheimer's disease-related dementias is vascular or Alzheimer dementia.
27. The method of claim 24, wherein said tauopatiry is selected from the group of argyrophilic grain dementia, corticobasal degeneration, dementia pugilistica, diffuse neurofibrillary tangles with calcification, frontotemporal dementia with parkinsonism, Prion-related disease, Hallervorden-Spatz disease, myotonic dystrophy, Niemann-Pick disease type C, non-Guamanian Motor Neuron disease with neurofibrillary tangles, Pick's disease, postencephalitic parkinsonism, prion protein cerebral amyloid angiopathy, progressive subcortical gliosis, progressive supranuclear palsy, subacute sclerosing panencephalitis, and tangle only dementia.
2 . The method of claim 24, wherein said α-synucleinopathy is selected from the group of dementia with Lewy bodies, multiple system atrophy with glial cytoplasmic inclusions, Shy-Drager syndrome, striatonigral degeneration, olivopontocerebellar atrophy, neurodegeneration with brain iron accumulation type I, olfactory dysfunction, and amyotrophic lateral sclerosis.
29. The method of claim 24, wherein said condition of the central or peripheral nervous system or systemic organ is Parkinson's disease.
30. The method of claim 29, wherein said Parkinson's disease is familial.
31. The method of claim 29, wherein said Parkinson's disease is non-familial
32. The method of claim 24, wherein said Motor Neuron Disease is associated with filaments and aggregates of neurofilament and/or superoxide dismutase proteins.
33. The method of claim 24, wherein said Spastic paraplegia is associated with defective function of chaperones and/or triple A proteins.
34. The method of claim 24, wherein said spinocerebellar ataxia is DRPLA or Machado- Joseph Disease.
35. The method of claim 24, wherein said Prion related disease is selected from the group of Creutzfeldt- Jakob disease, Gerstmann-Straussler-Scheinker disease, and variant Creutzfeldt- Jakob disease.
36. The method of claim 34, wherein said Amyloid Polyneuropathy is Senile amyloid polyneuropathy or Systemic Amyloidosis.
37. The method of claim 1 , wherein said compound is administered at a dose of about 1 mg to about 1 g per kg of the weight of said subject.
3δ. The method of claim 37, wherein said compound is administered at a dose of about 1 mg to about 200 mg per kg of the weight of said subject.
39. The method of claim 37, wherein said compound is administered at a dose of about 10 mg to about 100 mg per kg of the weight of said subject.
40. The method of claim 37, wherein said compound is administered at a dose of about 30 mg to about 70 mg per kg of the weight of said subject.
41. A method of preventing abnormal protein folding, abnormal protein aggregation, amyloid formation, deposition, accumulation, or persistence, or amyloid lipid interactions in a subject comprising administering to said subject a pharmaceutically effective amount of a compound having the following structure:
Figure imgf000044_0001
wherein each of R„ R,,, R2, R2,, R3, R3,, R4, R4,, R5, R5 ,, R6, and R6, is independently selected from the group of:
(a) hydrogen atom; (b) NHR7, wherein said R7 is selected from the group ofhydrogen; C2-C10 acyl and C1-C10 alkyl; (c) NR R9, wherein said Rg is C2-C10 acyl or Ci-Cjo alkyl and said R9 is
C2-C10 acyl or Ci -C10 alkyl;
(d) ORJOJ wherein said R^ø is selected from the group of no group, hydrogen, C2-Cιo acyl, C i-Cio alkyl and SO3H;
(e) C5-C7 glycosyl; (f) C3-Cg cycloalkyl optionally substituted with a substituent selected from the group ofhydrogen, OH, NH2, SH, OSO3H and OPO3H2; (g) SRi 1 , wherein Ri 1 is selected from the group of hydrogen, C 1 -C 10 alkyl and O3H; (h) C 1 -C j 0 alkyl optionally substituted with a substituent selected from the group of hydrogen, OR i0i. NHR7, NR R9 and SRj j; and
(i) C3-C cycloalkyl optionally substituted with a substituent selected from the group of hydrogen,. ORJQ, NHR7, NR R9 and SRi 1 providing that the compound is not myo-inositol.
42. The method of claim 41, wherein said compound is said compound is a 1,2,3,4,5,6-cyclohexanehexol.
43. The method of claim 42, wherein said compunds is selected from the group of cis-, epi-, allo-, muco-, neo-, scyllo-, O-chiro- and L-c/iZro-inositols.
44. The method of claim 41, wherein said compound is a 1,2,3,4,5- cyclohexanepentol.
45. The method of claim 44, wherein said compound is a quercitol selected from the group of epi-, vibo-, scyllo-, allo-, talo-, gala-, cis-, muco-, neo-, proto- quercitols and enantiomers thereof.
46. The method of claim 41 , wherein said compound is selected from the group of a cyclohexanetetrol, a cyclohexanetriol, stereoisomer of cyclohexanetetrol, stereoisimer of cyclohexanetriol, enantiomer of cyclohexanetetrol, and enantiomer of cyclohexanetriol.
47. The method of claim 41, wherein said compound is a pentahydroxycyclohexanone or a stereoisomer or an enantoimer thereof.
4δ. The method of claim 47, wherein said compound is an inosose selected from the group of scy/Zo-inosose, L-c/zz'ro-inosose-1 and L-epz'-inosose.
49. The method of claim 41, wherein said compound is a trihydroxycyclohexanone, or a stereoisomer or an enantiomer thereof.
50. The method of claim 49, wherein said compound is (-)-l-rfeo y-scyllo- inosose.
51. The method of claim 41 , wherein said compound is a pentahydroxycyclohexanone, or a stereoisomer or an enantiomer thereof.
52. The method of claim 51, wherein said compound is an inosose selected from the group of sςyZZo-inososβ, L-c/zzVo-inosose-1 and L-epz'-inosose.
53. The method of claim 41 , wherein said compound is a trihydroxyxcyclohexanone or a stereoisomer or an enantiomer thereof.
54. The method of claim 53, wherein said compound is (-)-l- reo y-_?cyZZo- inosose.
55. The method of claim 41 , wherein said compound is an O-monomethyl- cyclohexanehexol or a stereoisomer or an enantiomer thereof.
56. The method of claim 55, wherein said compound is selected from the group of D-pinitol, L-quebrachitol and D-bornesitol.
57. The method of claim 41, wherein said compound is selected from the group of monoaminocyclohexanepentols (inosamines), diaminocyclohexanetetrols (inosadiamines), diaminocyclohexanetriols, tstereoisomers thereof and, enantiomers thereof, and pharmaceutically acceptable salts thereof.
58. . The method of claim 57, wherein said compound is selected from the group of L-neo-inosamine, DL-epz-inosamine-2, streptamine and deoxystreptamine.
59. The method of claim 41 , wherein said compound is a monomercapto- cyclohexanepentol or a stereoisomer or an enantiomer thereof
60. The method of claim 59, wherein said compound is T -l-deoxy-1- mercapto-8-O-methyl-chiro-inositol.
61. The method of claim 41, wherein said compound is scyZZo-inositol.
62. The method of claim 41, wherein said compound is «//o-inositol.
63. The method of claim 41, wherein said condition of the central or
aasmuTE SHEET <røtf peripheral nervous system or systemic organ results in the deposition of proteins, protein fragments and peptides in beta-pleated sheats and/or fibrils and/or aggregates.
64. The method of claim 63, wherein said condition of the central or peripheral nervous system or systemic organ is selected from the group of: Alzheimer's disease, presenile and senile forms; amyloid angiopathy; mild cognitive impairment; Alzheimer's disease-related dementia; tauopathy; α-synucleinopathy; Parkinson's disease; Amyotrophic Lateral Sclerosis; motor neuron Disease; Spastic paraplagia; Huntington's Disease, spinocerebellar ataxia, Freidrich's Ataxia; neurodegenerative diseases associated with intracellular and/or intraneuronal aggregates of proteins with polyglutamine, polyalanine or other repeats arising from pathological expansions of tri- or tetra-nucleotide elements within corresponding genes; cerebrovascular diseases; Down's syndrome; head trauma with post-traumatic accumulation of amyloid beta peptide; Prion related disease; Familial British Dementia; Familial Danish Dementia; Presenile Dementia with Spastic Ataxia; Cerebral Amyloid Angiopathy, British Type; Presenile Dementia With Spastic Ataxia Cerebral Amyloid Angiopathy, Danish Type; Familial encephalopathy with neuroserpin inclusion bodies (FENIB); Amyloid Polyneuropathy; Inclusion Body myositis due to amyloid beta peptide; Familial and Finnish Type Amyloidosis; Systemic amyloidosis associated with multiple myeloma; Familial Mediterranean Fever; chronic infections and inflammations; and Type JJ Diabetes Mellitus associate with islet amyloid polypeptide (IAPP).
65. The method of claim 64, wherein said condition of the central or peripheral nervous system or systemic organ is Alzheimer's disease.
66. The method of claim 64, wherein said Alzheimer's disease-related dementias is vascular or Alzheimer dementia.
67. The method of claim 64, wherein said tauopathy is selected from the group of argyrophilic grain dementia, corticobasal degeneration, dementia pugilistica, diffuse neurofibrillary tangles with calcification, frontotemporal dementia with parkinsonism, Prion-related disease, Hallervorden-Spatz disease, myotonic dystrophy, Niemann-Pick disease type C, non-Guamanian Motor Neuron disease with neurofibrillary tangles, Pick's disease, postencephalitic parkinsonism, prion protein cerebral amyloid angiopathy, progressive subcortical gliosis, progressive supranuclear palsy, subacute sclerosing panencephalitis, and tangle only dementia.
68. The method of claim 64, wherein said α-synucleinopathy is selected from the group of dementia with Lewy bodies, multiple system atrophy with glial cytoplasmic inclusions, Shy-Drager syndrome, striatonigral degeneration, olivopontocerebellar atrophy, neurodegeneration with brain iron accumulation type I, olfactory dysfunction, and amyotrophic lateral sclerosis.
69. The method of claim 24, wherein said condition of the central or peripheral nervous system or systemic organ is Parkinson's disease.
70. The method of claim 69, wherein said Parkinson's disease is familial.
71. The method of claim 69, wherein said Parkinson's disease is non-familial
72. The method of claim 64, wherein said Motor Neuron Disease is associated with filaments and aggregates of neuro filament and/or superoxide dismutase proteins.
73. The method of claim 64, wherein said Spastic paraplagia is associated with defective function of chaperones and/or triple A proteins.
74. The method of claim 64, wherein said spinocerebellar ataxia is DRPLA or Machado-Joseph Disease.
75. The method of claim 64, wherein said Prion related disease is selected from the group of Creutzfeldt- Jakob disease, Gerstmann-Straussler-Scheinker disease, and variant Creutzfeldt-Jakob disease.
76. The method of claim 64, wherein said Amyloid Polyneuropathy is Senile amyloid polyneuropathy or Systemic Amyloidosis.
77. The method of claim 41, wherein said compound is administered at a dose of about 1 mg to about 1 g per kg of the weight of said subject.
78. The method of claim 77, wherein said compound is administered at a dose of about 1 mg to about 200 mg per kg of the weight of said subject.
79. The method of claim 77, wherein said compound is administered at a dose of about 10 mg to about 100 mg per kg of the weight of said subject.
80. The method of claim 77, wherein said compound is administered at a dose of about 30 mg to about 70 mg per kg of the weight of said subject.
δ 1. A method of causing the dissociation of abnormally aggregated proteins and/or dissolving or disrupting pre-formed or pre-deposited amyloid fibril or amyloid in a subject comprising administering to said subject a pharmaceutically effective amount of a compound having the following structure:
Figure imgf000051_0001
wherein each of R1} R , R2, R2., R3, R3., R4, R4-, R5, R5 •, Re, and R6 , is independently selected from the group of:
(a) hydrogen atom;
(b) NHR7, wherein said R7 is selected from the group ofhydrogen; C2-C1 Q acyl and CI~C Q alkyl; (c) NR R9, wherein said Rg is C2-C1 Q acyl or CJ-CJO alkyl and said R9 is
C2-C10 acyl or CJ-CJO alkyl;
(d) ORJO» wherein said R^o is selected from the group of no group, hydrogen, C2-C10 acyl, C 1-CJ O alkyl and SO3H;
(e) C5-C7 glycosyl; (f) C3-Cg cycloalkyl optionally substituted with a substituent selected from the group ofhydrogen, OH, NH2, SH, OSO3H and OPO3H2; (g) SRi 1, wherein R\ \ is selected from the group ofhydrogen, Cj-Cio alkyl and O3H; (h) CJ-CJO alkyl optionally substituted with a substituent selected from the group of hydrogen, OR Q, NHR7, NR R9 and SRi \ and
(i) C3-Cg cycloalkyl optionally substituted with a substituent selected from the group ofhydrogen, ORJQ, NHR7, NR R9 and SR\ \ providing that the compound is not myo-inositol.
82. The method of claim 81, wherein said compound is said compound is a 1 ,2,3,4,5,6-cyclohexanehexol.
83. The method of claim 82, wherein said compunds is selected from the group of cis-, epi-, allo-, muco-, neo- , scyllo-, O-chiro- and L-c/zz'rσ-inositols.
δ4. The method of claim δl, wherein said compound is a 1,2,3 ,4,5- cyclohexanepentol.
δ5. The method of claim δ4, wherein said compound is a quercitol selected from the group of epi-, vibo-, scyllo-, allo-, talo-, gala-, cis-, muco-, neo-, proto- quercitols and enantiomers thereof.
86. The method of claim 81 , wherein said compound is selected from the group of a cyclohexanetetrol, a cyclohexanetriol, stereoisomer of cyclohexanetetrol, stereoisimer of cyclohexanetriol, enantiomer of cyclohexanetetrol, and enantiomer of cyclohexanetriol.
87. The method of claim 81, wherein said compound is a pentahydroxycyclohexanone or a stereoisomer or an enantoimer thereof.
8δ. The method of claim δ7, wherein said compound is an inosose selected from the group of scyZ/o-inosose, L-c/zzro-inosose-1 and L-epz'-inosose.
δ9. The method of claim 81, wherein said compound is a trihydroxycyclohexanone, or a stereoisomer or an enantiomer thereof.
90. The method of claim 89, wherein said compound is (-)-l-deoxy-scyllo- inosose.
91. The method of claim 81 , wherein said compound is a pentahydroxycyclohexanone, or a stereoisomer or an enantiomer thereof.
92. The method of claim 91, wherein said compound is an inosose selected from the group of scy Z/o-inosose, L-c/πro-inosose-1 and L-epz'-inosose.
93. The method of claim 81 , wherein said compound is a trihydroxyxcyclohexanone or a stereoisomer or an enantiomer thereof.
94. The method of claim 93, wherein said compound is (-)-l-deoxy-scyllo- inosose.
95. The method of claim .81 , wherein said compound is an O-monomethyl- cyclohexanehexol or a stereoisomer or an enantiomer thereof.
96. The method of claim 95, wherein said compound is selected from the group of D-pinitol, L-quebrachitol and D-bornesitol.
97. The method of claim 81, wherein said compound is selected from the group of monoaminocyclohexanepentols (inosamines), diaminocyclohexanetetrols (inosadiamines), diaminocyclohexanetriols, tstereoisomers thereofand, enantiomers thereof, and pharmaceutically acceptable salts thereof.
98. The method of claim 97, wherein said compound is selected from the group of L-neo-inosamine, DL-ejpz'-inosamine-2, streptamine and deoxystreptamine.
99. The method of claim 81 , wherein said compound is a monomercapto- cyclohexanepentol or a stereoisomer or an enantiomer thereof.
100. The method of claim 99, wherein said compound is IL-l-deoxy-l- mercaρto-8-O-methyl-chiro-inositol.
101. The method of claim 91 , wherein said compound is scyZZo-inositol.
102. The method of claim 81 , wherein said compound is Z/o-inositol.
103. The method of claim 81 , wherein said condition of the central or peripheral nervous system or systemic organ results in the deposition of proteins, protein fragments and peptides in beta-pleated sheats and/or fibrils and/or aggregates.
104. The method of claim 103, wherein said condition of the central or peripheral nervous system or systemic organ is selected from the group of: Alzheimer's disease, presenile and senile forms; amyloid angiopathy; mild cognitive impairment; Alzheimer's disease-related dementia; tauopathy; α-synucleinopathy; Parkinson's disease; Amyotrophic Lateral Sclerosis; motor neuron Disease; Spastic paraplagia; Huntington's Disease, spinocerebellar ataxia, Freidrich's Ataxia; neurodegenerative diseases associated with intracellular and/or intraneuronal aggregates of proteins with polyglutamine, polyalanine or other repeats arising from pathological expansions of tri- or tetra-nucleotide elements within corresponding genes; cerebrovascular diseases; Down's syndrome; head trauma with post-traumatic accumulation of amyloid beta peptide; Prion related disease; Familial British Dementia; Familial Danish Dementia; Presenile Dementia with Spastic Ataxia; Cerebral Amyloid Angiopathy, British Type; Presenile Dementia With Spastic Ataxia Cerebral Amyloid Angiopathy, Danish Type; Familial encephalopathy with neuroserpin inclusion bodies (FENIB); Amyloid Polyneuropathy; Inclusion Body myositis due to amyloid beta peptide; Familial and Finnish Type Amyloidosis; Systemic amyloidosis associated with multiple myeloma; Familial Mediterranean Fever; chronic infections and inflammations; and Type JJ Diabetes Mellitus associate with islet amyloid polypeptide (IAPP).
105. The method of claim 104, wherein said condition of the central or peripheral nervous system or systemic organ is Alzheimer's disease.
106. The method of claim 104, wherein said Alzheimer's disease-related dementias is vascular or Alzheimer dementia.
107. The method of claim 104, wherein said tauopathy is selected from the group of argyrophilic grain dementia, corticobasal degeneration, dementia pugilistica, diffuse neurofibrillary tangles with calcification, frontotemporal dementia with parkinsonism, Prion-related disease, Hallervorden-Spatz disease, myotonic dystrophy, Niemann-Pick disease type C, non-Guamanian Motor Neuron disease with neurofibrillary tangles, Pick's disease, postencephalitic parkinsonism, prion protein cerebral amyloid angiopathy, progressive subcortical gliosis, progressive supranuclear palsy, subacute sclerosing panencephalitis, and tangle only dementia.
108. The method of claim 104, wherein said α-synucleinopathy is selected from the group of dementia with Lewy bodies, multiple system atrophy with glial cytoplasmic inclusions, Shy-Drager syndrome, striatonigral degeneration, olivopontocerebellar atrophy, neurodegeneration with brain iron accumulation type I , olfactory dysfunction, and amyotrophic lateral sclerosis.
109. The method of claim 84, wherein said condition of the central or peripheral nervous system or systemic organ is Parkinson's disease.
110. The method of claim 109, wherein said Parkinson's disease is familial.
111. The method of claim 109, wherein said Parkinson's disease is non-familial
112. The method of claim 104, wherein said Motor Neuron Disease is associated with filaments and aggregates of neurofilament and/or superoxide dismutase proteins.
113. The method of claim 104, wherein said Spastic paraplagia is associated with defective function of chaperones and/or triple A proteins.
114. The method of claim 104, wherein said spinocerebellar ataxia is DRPLA or Machado-Joseph Disease.
115. The method of claim 104, wherein said Prion related disease is selected from the group of Creutzfeldt- Jakob disease, Gerstmann-Straussler-Scheinker disease, and variant Creutzfeldt- Jakob disease.
116. The method of claim 104, wherein said Amyloid Polyneuropathy is Senile amyloid polyneuropathy or Systemic Amyloidosis.
117. The method of claim 81 , wherein said compound is administered at a dose of about 1 mg to about 1 g per kg of the weight of said subject.
118. The method of claim 117, wherein said compound is administered at a dose of about 1 mg to about 200 mg per kg of the weight of said subject.
119. The method of claim 117, wherein said compound is administered at a dose of about 10 mg to about 100 mg per kg of the weight of said subject.
120. The method of claim 117, wherein said compound is administered at a dose of about 30 mg to about 70 mg per kg of the weight of said subject.
121. A method of diagnosing the presence of abnormally folded or aggregated protein and/or amyloid fibril or amyloid in a subject comprising:
(a) administering to said subject a radioactive compound or compound tagged with a substance that emits a detectable signal in a quantity sufficient and under conditions to allow for the binding of said compound to the abnormally folded or aggregated protein and/or fibrils or amyloid, if present; and
(b) detecting the radioactivity or the signal from the compound bound to the abnormally folded or aggregated protein and/or fibrils or amyloid, thus diagnosing the presence of abnormally folded or aggregated protein and/or amyloid fibril or amyloid in said subject, wherein said compound has the following structure:
Figure imgf000057_0001
wherein each of Rl5 R , R2, R2,, R3, R3,, R4, R4,, R5, R5 ,, R6, and R6, is independently selected from the group of:
(a) hydrogen atom;
(b) NHR7, wherein said R7 is selected from the group ofhydrogen; C2-C10 acyl and C -C Q alkyl;
(c) NRgR9, wherein said Rg is C2-C10 acyl or Cj-Cio alkyl and said R9 is C2-C10 acyl or Cj-Cio alkyl;
(d) ORJO, wherein said Rio is selected from the group of no group, hydrogen, C2-C10 acyl, C i-Cio alkyl and SO3H; (e) C5-C7 glycosyl;
(f) C3-C cycloalkyl optionally substituted with a substituent selected from the group ofhydrogen, OH, NH2, SH, OSO3H and OPO3H2;
(g) SRj \, wherein R \ is selected from the group ofhydrogen, CJ-CJO alkyl and O3H; (h) Ci-CjQ a kyl optionally substituted with a substituent selected from the group of hydrogen, ORjrj, NHR7> NR R9 and SRj \; and (i) C3~Cg cycloalkyl optionally substituted with a substituent selected from the group ofhydrogen, ORJQ, NHR7, NR R9 and SRj providing that the compound is not myo-inositol.
122. The method of claim 121, wherein said compound is said compound is a 1 ,2,3,4,5,6-cyclohexanehβxol.
123. The method of claim 122, wherein said compounds is selected from the group of cis-, epi-, olio-, muco-, neo- , scyllo-, O-chiro- and L-cArro-inositols.
124. The method of claim 121, wherein said compound is a 1,2,3,4,5- cyclohexanepentol.
125. The method of claim 124, wherein said compound is quercitol selected from the group of epz'-, vibo-, scyllo-, αllo-, tαlo-, gαlα-, cis-, muco-, neo-, proto- quercitols and enantiomers thereof.
126. The method of claim 121, wherein said compound is selected from the group of a cyclohexanetetrol, a cyclohexanetriol, stereoisomer of cyclohexanetetrol, stereoisimer of cyclohexanetriol, enantiomer of cyclohexanetetrol, and enantiomer of cyclohexanetriol.
127. The method of claim 121, wherein said compound is a pentahydroxycyclohexanone or a stereoisomer or an enantoimer thereof.
128. The method of claim 127, wherein said compound is an inosose selected from the group of scy/Zo-inosose, L-cAzVo-inosose-1 and L-epz'-inosose.
129. The method of claim 121, wherein said compound is a trihydroxyxcyclohexanone, or a stereoisomer or an enantiomer thereof.
130. The method of claim 129, wherein said compound is (-)-l -deoxy-scy lo- inosose.
131. The method of claim 121, wherein said compound is a pentahydroxycyclohexanone, or a stereoisomer or an enantiomer thereof.
132. The method of claim 131, wherein said compound is an inosose selected from the group of ,$cy//o-inosose, L-c/w'ro-inosose-1 and L-e/?z-inosose.
133. The method of claim 121, wherein said compound is a trihydroxyxcyclohexanone or a stereoisomer or an enantiomer thereof.
134. The method of claim 133, wherein said compound is (-)-l-deoxy-scyllo- inosose.
135. The method of claim 121, wherein said compound is an O-monomethyl- cyclohexanehexol or a stereoisomer or an enantiomer thereof.
136. The method of claim 135, wherein said compound is selected from the group of D-pinitol, L-quebrachitol and D-bomesitol.
137. The method of claim 121, wherein said compound is selected from the group of monoaminocyclohexanepentols (inosamines), diaminocyclohexanetetrols (inosadiamines), diaminocyclohexanetriols, tstereoisomers thereofand, enantiomers thereof, and pharmaceutically acceptable salts thereof.
138. The method of claim 137, wherein said compound is selected from the group of L-weo-inosamine, DL-epz-inosamine-2, streptamine and deoxystreptamine.
139. The method of claim 121, wherein said compound is a monomercapto- cyclohexanepentol or a stereoisomer or an enantiomer thereof
140. The method of claim 139, wherein said compound is X -l-deox)?-\- mercaρto-8-O-methyl-chiro-inositol.
141. The method of claim 121, wherein said compound is scy/Zo-inositol.
142. The method of claim 121, wherein said compound is ZZo-inositol.
143. The method of claim 121, wherein said condition of the central or peripheral nervous system or systemic organ results in the deposition of proteins, protein fragments and peptides in beta-pleated sheats and/or fibrils and/or aggregates.
144. The method of claim 143, wherein said condition of the central or peripheral nervous system or systemic organ is selected from the group of: Alzheimer's disease, presenile and senile forms; amyloid angiopathy; mild cognitive impairment; Alzheimer's disease-related dementia; tauopathy; α-synucleinopathy; Parkinson's disease; Amyotrophic Lateral Sclerosis; motor neuron Disease; Spastic paraplagia; Huntington's Disease, spinocerebellar ataxia, Freidrich's Ataxia; neurodegenerative diseases associated with intracellular and/or intraneuronal aggregates of proteins with polyglutamine, polyalanine or other repeats arising from pathological expansions of tri- or tetra-nucleotide elements within corresponding genes; cerebrovascular diseases; Down's syndrome; head trauma with post-traumatic accumulation of amyloid beta peptide; Prion related disease; Familial British Dementia; Familial Danish Dementia; Presenile Dementia with Spastic Ataxia; Cerebral Amyloid Angiopathy, British Type; Presenile Dementia With Spastic Ataxia Cerebral Amyloid Angiopathy, Danish Type; Familial encephalopathy with nβuroserpin inclusion bodies (FENIB); Amyloid Polyneuropathy; Inclusion Body myositis due to amyloid beta peptide; Familial and Finnish Type Amyloidosis; Systemic amyloidosis associated with multiple myeloma; Familial Mediterranean Fever; chronic infections and inflammations; and Type π Diabetes Mellitus associate with islet amyloid polypeptide (IAPP).
145. The method of claim 144, wherein said condition of the central or peripheral nervous system or systemic organ is Alzheimer's disease.
146. The method of claim 144, wherein said Alzheimer's disease-related dementias is vascular or Alzheimer dementia.
147. The method of claim 144, wherein said tauopathy is selected from the group of argyrophilic grain dementia, corticobasal degeneration, dementia pugilistica, diffuse neurofibrillary tangles with calcification, frontotemporal dementia with parkinsonism, Prion-related disease, Hallervorden-Spatz disease, myotonic dystrophy, Niemann-Pick disease type C, non-Guamanian Motor Neuron disease with neurofibrillary tangles, Pick's disease, postencephalitic parkinsonism, prion protein cerebral amyloid angiopathy, progressive subcortical gliosis, progressive supranuclear palsy, subacute sclerosing panencephalitis, and tangle only dementia.
148. The method of claim 144, wherein said α-synucleinopathy is selected from the group of dementia with Lewy bodies, multiple system atrophy with glial cytoplasmic inclusions, Shy-Drager syndrome, striatonigral degeneration, olivopontocerebellar afrophy, neurodegeneration with brain iron accumulation type I , olfactory dysfunction, and amyotrophic lateral sclerosis.
149. The method of claim 144, wherein said condition of the central or peripheral nervous system or systemic organ is Parkinson's disease.
150. The method of claim 149, wherein said Parkinson's disease is familial.
151. The method of claim 149, wherein said Parkinson's disease is non-familial
152. The method of claim 144, wherein said Motor Neuron Disease is associated with filaments and aggregates of neurofilament and/or superoxide dismutase proteins.
153. The method of claim 144, wherein said Spastic paraplagia is associated with defective function of chaperones and/or triple A proteins.
154. The method of claim 144, wherein said spinocerebellar ataxia is DRPLA or Machado- Joseph Disease.
155. The method of claim 144, wherein said Prion related disease is selected from the group of Creutzfeldt- Jakob disease, Gerstmann-Straussler-Scheinker disease, and variant Creutzfeldt-Jakob disease.
156. The method of claim 144, wherein said Amyloid Polyneuropathy is Senile amyloid polyneuropathy or Systemic Amyloidosis.
157. The method of claim 121, wherein said compound is administered at a dose of about 1 mg to about 1 g per kg of the weight of said subject.
158. The method of claim 157, wherein said compound is administered at a dose of about 1 mg to about 200 mg per kg of the weight of said subject.
159. The method of claim 121, wherein said detectable signal is a fluorescent signal.
160. The method of claim 121, wherein said detectable signal is a radioactive signal.
161. A method of diagnosing the presence of abnormally folded or aggregated protein and/or amyloid fibril or amyloid in a subject comprising:
(a) collecting a sample from said subject;
(b) contacting said sample with a radioactive compound or compound tagged with a substance that emits a detectable signal under conditions to allow the binding of said compound to the abnormally folded or aggregated protein and/or amyloid fibril or amyloid if present; and
(c) detecting the radioactivity or the signal from the compound bound to the abnormally folded or aggregated protein and/or fibrils or amyloid, thus diagnosing the presence of abnormally folded or aggregated protein and/or amyloid fibril or amyloid in said subject, wherein said compound has the following structure:
Figure imgf000064_0001
wherein each of R„ R,,, R2, R2,, R3, R3., R , R4-5 R5, R5 >, R6, and R6. is independently selected from the group of: (a) hydrogen atom; (b) NHR7, wherein said R7 is selected from the group ofhydrogen; C2-C10 acyl and CJ-CIQ alkyl;
(c) NRgRp, wherein said Rg is C2-C1 ø acyl or Ci -C\ Q alkyl and said R9 is C2-C10 acyl or CJ-CJO alkyl;
(d) OR jo, wherein said JQ is selected from the group of no group, hydrogen, C2-C10 acyl, C 1-C10 alkyl and SO3H;
(e) C5-C7 glycosyl;
(f) C3~Cg cycloalkyl optionally substituted with a substituent selected from the group ofhydrogen, OH, NH2, SH, OSO3H and OPO3H2;
(g) SR] 1 , wherein R\ \ is selected from the group of hydrogen, C \ -C \ g alkyl and O3H;
(h) C\-C Q alkyl optionally substituted with a substituent selected from the group of hydrogen, ORIQ, NHRJ, NR R9 and SRn; and (i) C3-Cg cycloalkyl optionally substituted with a substituent selected from the group ofhydrogen, OR Q, NHR7, NRgR9 and SR^ j ? providing that the compound is not myo-inositol.
162. The method of claim 161, wherein said compound is said compound is a 1 ,2,3,4,5,6-cyclohexanehexol.
163. The method of claim 162, wherein said compunds is selected from the group of cis-, epi-, allo-, muco-, neo-, scyllo-, O-chiro- and L-e/zzVo-inositols.
164. The method of claim 161, wherein said compound is a 1 ,2,3,4,5- cyclohexanepentol.
165. The method of claim 164, wherein said compound is a quercitol selected from the group of epi-, vibo-, scyllo-, allo-, talo-, gala-, cis-, muco-, neo-, proto- quercitols and enantiomers thereof.
166. The method of claim 161, wherein said compound is selected from the group of a cyclohexanetetrol, a cyclohexanetriol, stereoisomer of cyclohexanetetrol, stereoisimer of cyclohexanetriol, enantiomer of cyclohexanetetrol, and enantiomer of cyclohexanetriol.
167. The method of claim 161, wherein said compound is a pentahydxycyclohexanonβ, a stereoisomer or an enantoimer thereof.
168. The method of claim 167, wherein said compound is an inosose selected from the group of scy/Zo-inosose, L-c/zz'ro-inosose-1 and L-epz'-inosose.
169. The method of claim 161, wherein said compound is a trihydroxyxcyclohexanone, or a stereoisomer or an enantiomer thereof.
170. The method of claim 169, wherein said compound is (-)- 1 - deoxy-scyllo- inosose.
171. The method of claim 161, wherein said compound is a pentahydroxycyclohexanone, or a stereoisomer or an enantiomer thereof.
172. The method of claim 171, wherein said compound is an inosose selected from the group of scyZZo-inosose, L-c/zz>o-inosose-l and L-epz'-inosose.
173. The method of claim 161, wherein said compound is a trihydroxyxcyclohexanone or a stereoisomer or an enantiomer thereof.
174. The method of claim 163 , wherein said compound is (-)- 1 -deoxy-scyllo- inosose.
175. The method of claim 161, wherein said compound is an O-monomethyl- cyclohexanehexol or a stereoisomer or an enantiomer thereof.
176. The method of claim 175, wherein said compound is selected from the group of D-pinitol, L-quebrachitol and D-bomesitol.
177. The method of claim 161, wherein said compound is selected from the group of monoaminocyclohexanepentols (inosamines), diaminocyclohexanetetrols (inosadiamines), diaminocyclohexanetriols, tstereoisomers thereofand, enantiomers thereof, and pharmaceutically acceptable salts thereof.
178. The method of claim 177, wherein said compound is selected from the group of L-weo-inosamine, DL-e^z-inosamine-2, streptamine and deoxystreptamine.
179. The method of claim 161, wherein said compound is a monomercapto- cyclohexanepentol or a stereoisomer or an enantiomer thereof
180. The method of claim 179, wherein said compound is \~ -\-deoxy-\- mercapto-δ-O-methyl-chiro-inositol.
181. The method of claim 161, wherein said compound is scyZ/o-inositol.
182. The method of claim 161, wherein said compound is //o-inositol.
183. The method of claim 161, wherein said condition of the central or peripheral nervous system or systemic organ results in the deposition of proteins, protein fragments and peptides in beta-pleated sheats and/or fibrils and/or aggregates.
184. The method of claim 183, wherein said condition of the central or peripheral nervous system or systemic organ is selected from the group of: Alzheimer's disease, presenile and senile forms; amyloid angiopathy; mild cognitive impairment; Alzheimer's disease-related dementia; tauopathy; α-synucleinopathy; Parkinson's disease; Amyotrophic Lateral Sclerosis; motor neuron Disease; Spastic paraplagia; Huntington's Disease, spinocerebellar ataxia, Freidrich's Ataxia; neurodegenerative diseases associated with intracellular and/or intraneuronal aggregates of proteins with polyglutamine, polyalanine or other repeats arising from pathological expansions of tri- or tetra-nucleotide elements within corresponding genes; cerebrovascular diseases; Down's syndrome; head trauma with post-traumatic accumulation of amyloid beta peptide; Prion related disease; Familial British Dementia; Familial Danish Dementia; Presenile Dementia with Spastic Ataxia; Cerebral Amyloid Angiopathy, British Type; Presenile Dementia With Spastic Ataxia Cerebral Amyloid Angiopathy, Danish Type; Familial encephalopathy with neuroserpin inclusion bodies (FENTB); Amyloid Polyneuropathy; Inclusion Body myositis due to amyloid beta peptide; Familial and Finnish Type Amyloidosis; Systemic amyloidosis associated with multiple myeloma; Familial Mediterranean Fever; chronic infections and inflammations; and Type TJ Diabetes Mellitus associate with islet amyloid polypeptide (IAPP).
185. The method of claim 1 δ4, wherein said condition of the central or peripheral nervous system or systemic organ is Alzheimer's disease.
1 δ6. The method of claim 184, wherein said Alzheimer's disease-related dementias is vascular or Alzheimer dementia.
187. The method of claim 184, wherein said tauopathy is selected from the group of argyrophilic grain dementia, corticobasal degeneration, dementia pugilistica, diffuse neurofibrillary tangles with calcification, frontotemporal dementia with parkinsonism, Prion-related disease, Hallervorden-Spatz disease, myotonic dystrophy, Niemann-Pick disease type C, non-Guamanian Motor Neuron disease with neurofibrillary tangles, Pick's disease, postencephalitic parkinsonism, prion protein cerebral amyloid angiopathy, progressive subcortical gliosis, progressive supranuclear palsy, subacute sclerosing panencephalitis, and tangle only dementia.
188. The method of claim 1 δ4, wherein said -synucleinopathy is selected from the group of dementia with Lewy bodies, multiple system atrophy with glial cytoplasmic inclusions, Shy-Drager syndrome, striatonigral degeneration, olivopontocerebellar atrophy, neurodegeneration with brain iron accumulation type I , olfactory dysfunction, and amyotrophic lateral sclerosis.
189. The method of claim 1 δ4, wherein said condition of the central or peripheral nervous system or systemic organ is Parkinson's disease.
190. The method of claim 189, wherein said Parkinson' s disease is familial.
191. The method of claim 189, wherein said Parkinson' s disease is non-familial
192. The method of claim 184, wherein said Motor Neuron Disease is associated with filaments and aggregates of neurofilament and/or superoxide dismutase proteins.
193. The method of claim 184, wherein said Spastic paraplagia is associated with defective function of chaperones and/or triple A proteins.
194. The method of claim 184, wherein said spinocerebellar ataxia is DRPLA or Machado- Joseph Disease.
195. The method of claim 184, wherein said Prion related disease is selected from the group of Creutzfeldt- akob disease, Gerstmann-Straussler-Scheinker disease, and variant Creutzfeldt- akob disease.
196. The method of claim 184, wherein said Amyloid Polyneuropathy is Senile amyloid polyneuropathy or Systemic Amyloidosis.
197. The method of claim 161, wherein said detectable signal is a fluorescent signal.
198. The method of claim 161, wherein said detectable signal is a radioactive signal.
199. The method of claim 161, wherein said detectable signal is an enzyme- linked immunosorbent assay signal
200. The method of claim 161, wherein said sample is whole blood.
201. The method of claim 161, wherein said sample is plasma.
PCT/CA2004/000272 2003-02-27 2004-02-27 Methods of preventing, treating and diagnosing disorders of protein aggregation WO2004075882A1 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
CA2516563A CA2516563C (en) 2003-02-27 2004-02-27 Methods of preventing, treating and diagnosing disorders of protein aggregation
US10/547,286 US8859628B2 (en) 2003-02-27 2004-02-27 Method for preventing, treating and diagnosing disorders of protein aggregation
AU2004216544A AU2004216544B2 (en) 2003-02-27 2004-02-27 Methods of preventing, treating and diagnosing disorders of protein aggregation
SI200431216T SI1608350T1 (en) 2003-02-27 2004-02-27 Methods of preventing, treating and diagnosing disorders of protein aggregation
JP2006501433A JP4999453B2 (en) 2003-02-27 2004-02-27 Methods for preventing, treating and diagnosing protein aggregation disorders
EP04715226A EP1608350B1 (en) 2003-02-27 2004-02-27 Methods of preventing, treating and diagnosing disorders of protein aggregation
DK04715226T DK1608350T3 (en) 2003-02-27 2004-02-27 Procedure for the prevention, treatment and diagnosis of disorders of protein aggregation
BRPI0407910-8A BRPI0407910A (en) 2003-02-27 2004-02-27 uses of compounds for the prevention, treatment or diagnosis in an individual of a protein aggregation disorder
DE602004021362T DE602004021362D1 (en) 2003-02-27 2004-02-27 METHOD FOR THE PREVENTION, TREATMENT AND DIAGNOSIS OF PROBLEM DISTURBANCES
AT04715226T ATE432694T1 (en) 2003-02-27 2004-02-27 METHOD FOR PREVENTING, TREATING AND DIAGNOSING PROTEIN AGGREGATION DISORDERS
IL170476A IL170476A (en) 2003-02-27 2005-08-24 Use of scyllo-inositol in the manufacture of a medicament for treatment or prevention of alzheimer's disease, dementia, cognitive impairment and use of the medicament for diagnosing the presence of abnormal protein
IL220065A IL220065A (en) 2003-02-27 2012-05-30 Use of scyllo-inositol in the preparation of a medicament for improving cognition in a patient with down's syndrome and for improving behavioural outcome in a subject with alzheimer's disease
US14/475,859 US20150031776A1 (en) 2003-02-27 2014-09-03 Method of preventing, treating and diagnosing disorders of protein aggregation

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US45136303P 2003-02-27 2003-02-27
US60/451,363 2003-02-27
US52095803P 2003-11-17 2003-11-17
US60/520,958 2003-11-17
US52353403P 2003-11-19 2003-11-19
US60/523,534 2003-11-19
US10/787,621 US7521481B2 (en) 2003-02-27 2004-02-26 Methods of preventing, treating and diagnosing disorders of protein aggregation
US10/787,621 2004-02-26

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US10/547,286 A-371-Of-International US8859628B2 (en) 2003-02-27 2004-02-27 Method for preventing, treating and diagnosing disorders of protein aggregation
US14/475,859 Division US20150031776A1 (en) 2003-02-27 2014-09-03 Method of preventing, treating and diagnosing disorders of protein aggregation

Publications (1)

Publication Number Publication Date
WO2004075882A1 true WO2004075882A1 (en) 2004-09-10

Family

ID=32931587

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2004/000272 WO2004075882A1 (en) 2003-02-27 2004-02-27 Methods of preventing, treating and diagnosing disorders of protein aggregation

Country Status (15)

Country Link
US (7) US7521481B2 (en)
EP (4) EP2153829B1 (en)
JP (4) JP4999453B2 (en)
AT (1) ATE432694T1 (en)
AU (2) AU2004216544B2 (en)
BR (1) BRPI0407910A (en)
CA (1) CA2516563C (en)
CY (2) CY1109338T1 (en)
DE (1) DE602004021362D1 (en)
DK (3) DK2153829T3 (en)
ES (3) ES2520047T3 (en)
IL (2) IL170476A (en)
PT (3) PT2153829E (en)
SI (3) SI2311445T1 (en)
WO (1) WO2004075882A1 (en)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006053428A1 (en) * 2004-11-17 2006-05-26 Joanne Mclaurin Compositions comprising scyllo-inositol derivatives and methods to treat disorders of protein aggregation
WO2007041855A1 (en) * 2005-10-13 2007-04-19 Waratah Pharmaceuticals, Inc. Scyllo-inositol derivatives and their use in the treatment of diseases characterised by abnormal protein folding or aggregation or amyloid formation, deposition, accumulation or persistence
WO2007101353A1 (en) * 2006-03-09 2007-09-13 Waratah Pharmaceuticals Inc. A cyclohexane polyalcohol formulation for treatment of disorders of protein aggregation
WO2007129221A3 (en) * 2006-02-17 2008-03-20 Joanne Mclaurin Compositions comprising an epi-inositol compounds and methods for treatment of disorders of protein aggregation
WO2008034244A1 (en) * 2006-09-21 2008-03-27 Waratah Pharmaceuticals Inc. The combination of a cyclohexanehexol and a nsaid for the treatment of neurodegenerative diseases
WO2008124930A1 (en) * 2007-04-12 2008-10-23 Joanne Mclaurin Use of cyclohexanehexol derivatives for the treatment of polyglutamine diseases
WO2010011012A1 (en) 2008-07-22 2010-01-28 Postech Academy-Industry Foundation Inositol and trehalose derivatives and pharmaceutical compositions for treating neurodegenerative diseases comprising the same
WO2010063701A2 (en) * 2008-12-02 2010-06-10 Ge Healthcare Limited In vivo imaging method
US7807175B2 (en) * 2005-12-12 2010-10-05 Ac Immune Sa Therapeutic vaccine
US20100292157A1 (en) * 2006-11-24 2010-11-18 Antonio Cruz Combination Treatments for Alzheimer's Disease and Similar Diseases
US20100331267A1 (en) * 2007-04-12 2010-12-30 Mclaurin Joanne Use of cyclohexanehexol derivatives in the treatment of alpha-synucleinopathies
WO2012051395A1 (en) 2010-10-13 2012-04-19 Elan Pharmaceuticals, Inc. Methods of synthesis of scyllitol and related compounds
WO2012173808A1 (en) * 2011-06-03 2012-12-20 Elan Pharmaceuticals, Inc. Scyllo-inositol for the treatment of behavioral and psychiatric disorders
US8859628B2 (en) 2003-02-27 2014-10-14 JoAnne McLaurin Method for preventing, treating and diagnosing disorders of protein aggregation
US9116157B2 (en) 2010-11-05 2015-08-25 Brandeis University Ice-cleaved alpha-synuclein as a biomarker
WO2016047823A1 (en) * 2014-09-25 2016-03-31 영남대학교 산학협력단 Composition for inhibiting cellular senescence containing melandrium firmum rohrbach extract or bornesitol separated from same as active ingredient
EP3231804A1 (en) * 2008-03-21 2017-10-18 The General Hospital Corporation Compounds and compositions for the detection and treatment of alzheimer's disease and related disorders
US10058625B2 (en) 2004-02-24 2018-08-28 The General Hospital Corporation Catalytic radiofluorination
US11844846B2 (en) 2018-05-16 2023-12-19 Emory University Styrylbenzothiazole derivatives and uses in imaging methods

Families Citing this family (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2003291063A1 (en) * 2002-11-19 2004-06-15 Board Of Trustees Operating Michigan State University Antioxidant and antimicrobial agents and methods of use thereof
US8790637B2 (en) 2003-06-27 2014-07-29 DePuy Synthes Products, LLC Repair and regeneration of ocular tissue using postpartum-derived cells
ES2597837T3 (en) 2003-06-27 2017-01-23 DePuy Synthes Products, Inc. Postpartum cells derived from placental tissue, and methods of manufacturing and using them
US9592258B2 (en) 2003-06-27 2017-03-14 DePuy Synthes Products, Inc. Treatment of neurological injury by administration of human umbilical cord tissue-derived cells
US9572840B2 (en) 2003-06-27 2017-02-21 DePuy Synthes Products, Inc. Regeneration and repair of neural tissue using postpartum-derived cells
WO2005009291A2 (en) 2003-07-23 2005-02-03 Synapse Biomedical, Inc. System and method for conditioning a diaphragm of a patient
US8257680B1 (en) 2004-02-24 2012-09-04 The General Hospital Corporation Catalytic radiofluorination
US8193250B2 (en) * 2004-10-22 2012-06-05 Mount Sinai School Of Medicine Compositions and methods for treating alzheimer's disease and related disorders and promoting a healthy nervous system
AU2006202209B2 (en) * 2005-05-27 2011-04-14 Lifescan, Inc. Amniotic fluid derived cells
US9050005B2 (en) 2005-08-25 2015-06-09 Synapse Biomedical, Inc. Method and apparatus for transgastric neurostimulation
EP1996284A2 (en) 2006-03-09 2008-12-03 Synapse Biomedical, Inc. Ventilatory assist system and method to improve respiratory function
AU2007265631B2 (en) * 2006-06-23 2012-11-08 The Feinstein Institute For Medical Research Inhibitors of A-Beta and synuclein aggregation
US20080097153A1 (en) * 2006-08-24 2008-04-24 Ignagni Anthony R Method and apparatus for grasping an abdominal wall
US20110218176A1 (en) 2006-11-01 2011-09-08 Barbara Brooke Jennings-Spring Compounds, methods, and treatments for abnormal signaling pathways for prenatal and postnatal development
US20090214474A1 (en) * 2006-11-01 2009-08-27 Barbara Brooke Jennings Compounds, methods, and treatments for abnormal signaling pathways for prenatal and postnatal development
US20080103116A1 (en) * 2006-11-01 2008-05-01 Jennings-Spring Barbara L Method of treatment and compositions of D-chiro inositol and phosphates thereof
US9079016B2 (en) * 2007-02-05 2015-07-14 Synapse Biomedical, Inc. Removable intramuscular electrode
EP2121633A2 (en) 2007-02-12 2009-11-25 Merck & Co., Inc. Piperazine derivatives for treatment of ad and related conditions
SI2148667T1 (en) * 2007-04-12 2013-12-31 Waratah Pharmaceuticals, Inc. Use of cyclohexanehexol derivatives in the treatment of ocular diseases
WO2008124931A1 (en) * 2007-04-12 2008-10-23 Joanne Mclaurin Use of cyclohexanehexol derivatives in the treatment of amyotrophic lateral sclerosis
WO2008144578A1 (en) * 2007-05-17 2008-11-27 Synapse Biomedical, Inc. Devices and methods for assessing motor point electromyogram as a biomarker
US8428726B2 (en) 2007-10-30 2013-04-23 Synapse Biomedical, Inc. Device and method of neuromodulation to effect a functionally restorative adaption of the neuromuscular system
WO2009059033A1 (en) * 2007-10-30 2009-05-07 Synapse Biomedical, Inc. Method of improving sleep disordered breathing
US20090131857A1 (en) * 2007-11-20 2009-05-21 Mark Geiger Method and apparatus for introducing a medicinal dose directly into a mammalian patient's cerebrospinal fluid
AT506535B1 (en) * 2008-02-22 2010-04-15 Affiris Forschungs & Entwicklungs Gmbh VACCINE CONTAINING ALPHA SYNUCLEIN MIMOTOPES BASED ON PEPTIDES
WO2010040232A1 (en) * 2008-10-09 2010-04-15 Waratah Pharmaceuticals Inc. Use of scyllo-inositols for the treatment of macular degeneration-related disorders
BRPI1013409A2 (en) 2009-03-26 2018-01-16 Advanced Tech And Regenerative Medicine Llc human umbilical cord tissue cells as therapy for alzheimer's disease
US20100249073A1 (en) * 2009-03-27 2010-09-30 Robert Sabin Prophylactic and therapeutic treatment of Alzheimer's disease using phytic acid and phytate to reduce amyloid beta plaque and tau protein
WO2011100670A1 (en) * 2010-02-15 2011-08-18 Abbott Laboratories Process for the preparation of scyllo-inositol
KR101365711B1 (en) * 2013-04-30 2014-02-21 재단법인 경북해양바이오산업연구원 A new biosurfactant isolated from yeast
US10377855B2 (en) 2014-06-25 2019-08-13 Rensselaer Polytechnic Institute Oxetane polymers and methods of preparation thereof
US11191735B2 (en) 2015-03-13 2021-12-07 Nutrition 21, Llc Arginine silicate for periodontal disease
US20170135969A1 (en) 2015-11-12 2017-05-18 Jds Therapeutics, Llc Topical arginine-silicate-inositol for wound healing
US20170354669A1 (en) * 2016-02-24 2017-12-14 The Trustees Of Columbia University In The City Of New York Disruption of the interaction between amyloid beta peptide and dietary lipids
MX2019002468A (en) 2016-09-01 2019-09-18 Jds Therapeutics Llc Magnesium biotinate compositions and methods of use.
US11883807B2 (en) 2017-04-11 2024-01-30 Colorado State University Research Foundation Functionalization of metal-organic frameworks
GB2594170A (en) * 2018-11-02 2021-10-20 Nutrition 21 Llc Compositions containing inositol-stabilized arginine silicate complexes and inositol for improving cognitive function in video gamers
US11471683B2 (en) 2019-01-29 2022-10-18 Synapse Biomedical, Inc. Systems and methods for treating sleep apnea using neuromodulation
EP3789018A1 (en) * 2019-09-09 2021-03-10 Servicio Andaluz De Salud Composition and methods for enhancing or promoting the secretion of ghrelin to promote a healthy metabolic aging
AU2020407071A1 (en) 2019-12-16 2022-07-07 Nutrition 21, Llc Methods of production of arginine-silicate complexes
BR112022013296A2 (en) * 2020-01-03 2022-09-06 Biosearch S A COMPOSITION FOR USE IN THE TREATMENT OF COGNITIVE DISORDERS
KR20230051237A (en) * 2020-09-17 2023-04-17 가부시끼가이샤 레조낙 autophagy activator
WO2024054415A1 (en) * 2022-09-07 2024-03-14 Eirgen Pharma, Ltd. A combination of scyllo-inositol and flavones

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4758430A (en) * 1987-01-21 1988-07-19 Robert Sabin Method of treatment of Alzheimer's disease using phytic acid
US4847082A (en) * 1987-01-21 1989-07-11 Robert Sabin Method of treatment of Alzheimer's disease using phytic acid
US5112814A (en) * 1990-10-24 1992-05-12 Robert Sabin Method of treatment of Parkinson's disease using phytic acid

Family Cites Families (256)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2088897A (en) * 1935-03-25 1937-08-03 Goss Printing Press Co Ltd Printing press
US2557381A (en) * 1946-08-09 1951-06-19 William C Huebner Multiple unit printing press
DE2234089C3 (en) * 1972-07-08 1975-01-23 Automatic Druckmaschinenfabrik Dr. W. Hinniger U. Soehne, 1000 Berlin Offset web-fed rotary printing press
GB1581233A (en) * 1976-06-02 1980-12-10 Drg Uk Ltd Printing press
US4201706A (en) * 1978-09-22 1980-05-06 Burton, Parsons & Company, Inc. Treatment of corneal edema
US4454151A (en) * 1982-03-22 1984-06-12 Syntex (U.S.A.) Inc. Use of pyrrolo pyrroles in treatment of ophthalmic diseases
US4515722A (en) 1982-03-30 1985-05-07 Merck & Co., Inc. Phosphatidyl inositol analogs useful as anti-inflammatory/analgesic agents
US4474806A (en) * 1982-05-10 1984-10-02 Merck & Co., Inc. Sulfonyl or carbonyl inositol derivatives useful as anti-inflammatory/analgesic agents
IT1164225B (en) 1983-05-13 1987-04-08 Anic Spa INVERTED ANALOGS OF PENTAPEPTIDE ENHANCING BRADICHINA BPP5A AND METHODS FOR THEIR PREPARATION
DE3405663A1 (en) 1984-02-17 1985-08-22 Merck Patent Gmbh, 6100 Darmstadt Process for the preparation of scyllo-inositol
US5128332A (en) 1984-10-23 1992-07-07 Perstorp Ab Method of treating cardiovascular diseases using inositoltrisphosphate
SE465951B (en) 1984-10-23 1991-11-25 Perstorp Ab ISOMER OF INOSITOL TRIPHOSPHATE PHARMACEUTICAL STATEMENTS FOR SALT FOR USE AS THERAPEUTIC OR PROPHYLACTIC AGENTS AND COMPOSITIONS THEREOF
US5330979A (en) 1984-10-23 1994-07-19 Perstorp Ab Method of treating renal disorders with inositoltriphosphate
US5545632A (en) 1984-10-23 1996-08-13 Perstorp Ab Method of treating retroviral disease
SE465305B (en) 1986-04-16 1991-08-26 Perstorp Ab USE OF INOSITOL PHOSPHATE FOR THE PREPARATION OF A MEDICINE
US5023248A (en) 1984-10-23 1991-06-11 Perstorp Ab Method of treating diabetes with inositol triphosphate
US4735902A (en) 1984-10-23 1988-04-05 Matti Siren Stabilized composition containing inositoltriphosphate
US5407924A (en) 1984-10-23 1995-04-18 Perstorp Ab Method of treating pain using inositol triphosphate
IL74497A (en) * 1985-03-05 1990-02-09 Proterra Ag Pharmaceutical compositions containing phenyl carbamate derivatives and certain phenyl carbamate derivatives
DE3610107A1 (en) * 1986-03-26 1987-10-08 Voith Gmbh J M ACTUATING DEVICE FOR ADJUSTING A ROLLER
US5057507A (en) 1986-04-16 1991-10-15 Perstorp Ab Method of alleviating bone damage with inositoltriphosphate
US5135923A (en) 1986-04-16 1992-08-04 Perstorp Ab Method of treating cardiovascular diseases
US4758420A (en) * 1986-07-14 1988-07-19 The Dow Chemical Company Solvent extraction of polychlorinated organic compounds from porous materials
US4952396A (en) 1986-11-19 1990-08-28 Linus Pauling Institute Of Science & Medicine Method of using phytic acid for inhibiting tumor growth
NL195004C (en) * 1987-03-04 2003-11-04 Novartis Ag Pharmaceutical preparation containing phenyl carbamate.
JPH067158Y2 (en) 1987-03-18 1994-02-23 株式会社藤井合金製作所 Waterproof structure of piping terminal
FI95572C (en) * 1987-06-22 1996-02-26 Eisai Co Ltd Process for the preparation of a medicament useful as a piperidine derivative or its pharmaceutical salt
DE3808142A1 (en) * 1988-03-11 1989-09-21 Goebel Gmbh Maschf STORAGE FACILITIES
DE3814752C1 (en) * 1988-04-30 1989-08-31 J.M. Voith Gmbh, 7920 Heidenheim, De
US5334618A (en) * 1991-04-04 1994-08-02 The Children's Medical Center Corporation Method of preventing NMDA receptor-mediated neuronal damage
JPH03102492A (en) 1989-06-12 1991-04-26 Mitsui Eng & Shipbuild Co Ltd Data transmission system for automatic vending machine and compound function type automatic vending machine
SE8904355D0 (en) 1989-12-21 1989-12-21 Perstorp Ab medicament
US5111814A (en) * 1990-07-06 1992-05-12 Thomas Jefferson University Laryngeal pacemaker
US5217959A (en) 1990-09-06 1993-06-08 Robert Sabin Method of treating multiple sclerosis with phytic acid
JP2984043B2 (en) 1990-09-18 1999-11-29 旭化成工業株式会社 Myo-inositol dehydrogenase and method for producing the same
SE9102068L (en) * 1991-07-03 1993-01-04 Perstorp Ab DERIVATIVES OF INOSITOL, COMPOSITIONS CONTAINING THESE AND USE THEREOF
JPH05192163A (en) 1991-09-30 1993-08-03 Suntory Ltd Inositol dehydrogenase gene
ATE128708T1 (en) 1991-12-18 1995-10-15 Schering Corp IMIDAZOLYLALKYL-PIPERAZINE AND -DIAZEPINE DERIVATIVES AS HISTAMINE H3 AGONISTS/ANTAGONISTS.
SE9200547L (en) 1992-02-25 1993-06-14 Perstorp Ab A PHARMACEUTICAL COMPOSITION WITH PREPARED BIO-ACCESSIBILITY FOR INOSITOL PHOSPHATE
JP3251976B2 (en) 1992-06-23 2002-01-28 旭化成株式会社 Substantially pure microorganism producing myo-inositol dehydrogenase
US5351616A (en) * 1992-08-13 1994-10-04 Man Roland Druckmaschinen Ag Rotary web printing machine, particularly for printing on thick or carton-type stock webs with replaceable plate cylinders
DE4229494A1 (en) 1992-09-04 1994-03-10 Basotherm Gmbh Medicines for topical use on the eye to treat increased intraocular pressure
ATE292131T1 (en) 1992-10-05 2005-04-15 Virginia Tech Intell Prop INTERMEDIATE PRODUCT FOR PRODUCING D-CHIRO-INOSOSE AND (+)-D-CHIRO-INOSITOL
US5643562A (en) 1993-03-29 1997-07-01 Queen's University Of Kingston Method for treating amyloidosis
US5840294A (en) 1993-03-29 1998-11-24 Queen's University At Kingston Method for treating amyloidosis
US5972328A (en) 1993-03-29 1999-10-26 Queen's University At Kingston Method for treating amyloidosis
DK0619369T3 (en) 1993-04-05 2003-10-06 Aveve Nv Phytate hydrolysis and enzyme composition for hydrolysis of phytate
DK0712827T3 (en) 1993-08-11 1999-09-20 Zaidan Hojin Biseibutsu Process for the preparation of D-chiro-inositol
US5412080A (en) 1993-08-25 1995-05-02 President And Fellow Of Harvard College Enterobactin compounds
SE502574C2 (en) 1994-01-25 1995-11-13 Perstorp Ab A pharmaceutical composition with improved bioavailability of inositol phosphate
DE4408025A1 (en) * 1994-03-10 1995-09-14 Koenig & Bauer Ag Printing unit for a multi-color web-fed rotary printing machine
DE4430693B4 (en) * 1994-08-30 2005-12-22 Man Roland Druckmaschinen Ag Drives for a web-fed rotary offset printing machine
US5550166A (en) 1995-03-17 1996-08-27 Ostlund; Richard E. Pinitol and derivatives thereof for the treatment of metabolic disorders
US5858326A (en) 1995-06-06 1999-01-12 Neurochem, Inc. Methods of increasing amyloid deposition
US6221645B1 (en) * 1995-06-07 2001-04-24 Elan Pharmaceuticals, Inc. β-secretase antibody
JP3630344B2 (en) 1995-11-17 2005-03-16 北興化学工業株式会社 Process for producing inositol stereoisomers
TW513409B (en) * 1996-06-07 2002-12-11 Eisai Co Ltd Polymorphs of donepezil hydrochloride
WO1997046526A1 (en) * 1996-06-07 1997-12-11 Eisai Co., Ltd. Stable polymorphs of donepezil (1-benzyl-4-[(5,6-dimethoxy-1-indanon)-2-yl]methylpiperidine) hydrochloride and process for production
US6232486B1 (en) 1996-06-11 2001-05-15 Nutrimed Biotech Molecular probes and modulators for PI-PLC and PI 3-kinase
TW506836B (en) * 1996-06-14 2002-10-21 Janssen Pharmaceutica Nv Fast-dissolving galanthamine hydrobromide tablet
US5880099A (en) 1996-09-20 1999-03-09 The Regents Of The University Of California Inositol polyphosphates and methods of using same
US5977078A (en) 1996-09-20 1999-11-02 The Regents Of The Univesity Of California Inositol polyphosphate derivatives and methods of using same
US20030068316A1 (en) 1997-02-05 2003-04-10 Klein William L. Anti-ADDL antibodies and uses thereof
US6218506B1 (en) 1997-02-05 2001-04-17 Northwestern University Amyloid β protein (globular assembly and uses thereof)
DE69832695T2 (en) 1997-03-10 2006-09-14 Loma Linda University Medical Center, Loma Linda USE OF R-ENANTIOMER NON-TEROIDAL INFLAMMATORY INHIBITION FOR THE PREVENTION OF ALZHEIMER'S DISEASE
US5998485A (en) 1997-06-16 1999-12-07 Cedars-Sinai Medical Center Method for modulating immune response with inositol
US6153603A (en) 1997-06-27 2000-11-28 Perstorp Ab Method of treating angiogenesis in tumor tissue
GB9801899D0 (en) 1998-01-29 1998-03-25 Univ London Neurotrophic properties of ipgs analogues
DE19805898C2 (en) * 1998-02-13 2003-09-18 Roland Man Druckmasch Printing unit for a web-fed rotary printing machine
US6720190B1 (en) * 1998-03-27 2004-04-13 Ole Hindsgaul Methods for screening compound libraries
US5981168A (en) * 1998-05-15 1999-11-09 The University Of British Columbia Method and composition for modulating amyloidosis
US6227110B1 (en) * 1998-06-23 2001-05-08 Heidelberger Druckmaschinen Ag Wet printing press with throw-off mechanism
US6310073B1 (en) 1998-07-28 2001-10-30 Queen's University At Kingston Methods and compositions to treat glycosaminoglycan-associated molecular interactions
AUPP589398A0 (en) 1998-09-14 1998-10-08 Walter And Eliza Hall Institute Of Medical Research, The Immunogenic compositions and uses thereof
DK1115874T3 (en) 1998-09-24 2009-04-20 Pharmacia & Upjohn Co Llc Alzheimer's disease secretase
AUPP675898A0 (en) 1998-10-27 1998-11-19 Walter And Eliza Hall Institute Of Medical Research, The A method of activating t cells and agents useful for same
US6221856B1 (en) 1999-02-03 2001-04-24 Inologic, Inc. Inositol derivatives for inhibiting superoxide anion production
GB2389113B (en) 1999-02-10 2004-02-04 Elan Pharm Inc B-secretase inhibitor
CN1390232A (en) * 1999-02-10 2003-01-08 艾兰制药公司 Human beta-secretase enzyme, inhibitor, and its composition and use
US6967196B1 (en) 1999-02-26 2005-11-22 Bristol-Myers Squibb Company Sulfonamide compounds and uses thereof
US6818430B1 (en) 1999-06-07 2004-11-16 Hokko Chemical Industry Co., Ltd. Process for producing L-epi-2-inosose and novel process for producing epi-inositol
WO2000077030A1 (en) 1999-06-15 2000-12-21 Elan Pharmaceuticals, Inc. Statine-derived tetrapeptide inhibitors of beta-secretase
US6864240B1 (en) * 1999-06-15 2005-03-08 Elan Pharmaceuticals, Inc. Dipeptide inhibitors of β-secretase
AU5994900A (en) 1999-07-09 2001-01-30 Isis Innovation Limited Compounds for inhibiting diseases and preparing cells for transplantation
DE19937805A1 (en) * 1999-08-10 2001-02-15 Roland Man Druckmasch Printing unit
DE19937796B4 (en) * 1999-08-10 2007-03-22 Man Roland Druckmaschinen Ag printing unit
DE19937803A1 (en) * 1999-08-10 2001-02-15 Roland Man Druckmasch Printing unit
US20020052311A1 (en) * 1999-09-03 2002-05-02 Beka Solomon Methods and compostions for the treatment and/or diagnosis of neurological diseases and disorders
US6329256B1 (en) 1999-09-24 2001-12-11 Advanced Micro Devices, Inc. Self-aligned damascene gate formation with low gate resistance
JP2001163810A (en) 1999-09-30 2001-06-19 Microbial Chem Res Found Method for new synthesis of 1d-chiro-inositol and intermediate
EP1221865B1 (en) 1999-10-18 2007-01-03 Muscletech Research and Development Inc. Food supplement for increasing lean mass and strength
US7728043B2 (en) * 1999-10-22 2010-06-01 Kim Darrick S H L Methods for treatment of beta-amyloid protein-induced ocular disease
CA2390376A1 (en) 1999-11-09 2001-05-17 William Leonard Scott .beta.-aminoacid compounds useful for inhibiting .beta.-amyloid peptide release and/or its synthesis
EP1235789A2 (en) 1999-11-09 2002-09-04 Eli Lilly And Company $g(b)-AMINOACID COMPOUNDS USEFUL FOR INHIBITING $g(b)-AMYLOID PEPTIDE RELEASE AND/OR ITS SYNTHESIS
DE19963944C1 (en) * 1999-12-31 2001-06-13 Koenig & Bauer Ag Method to adjust cylinders of printing machine; involves moving abutment to limit stroke of axle holder for adjustable cylinder at space from base plate and applying adjusting and holding forces
WO2001053255A1 (en) 2000-01-24 2001-07-26 Merck Sharp & Dohme Limited Gamma-secretase inhibitors
DE10008215B4 (en) * 2000-02-23 2013-03-28 Manroland Web Systems Gmbh Printing unit for a rotary printing press with cross slide
GB0005251D0 (en) * 2000-03-03 2000-04-26 Merck Sharp & Dohme Therapeutic compounds
US20030144255A1 (en) * 2000-03-06 2003-07-31 Bain Allen I Compositions for prevention and treatment of dementia
DE60124684T2 (en) 2000-03-20 2007-09-13 Merck Sharp & Dohme Ltd., Hoddesdon SULPHONAMIDO-SUBSTITUTED BROKEN BICYCLOALKYL DERIVATIVES
CA2401749A1 (en) 2000-03-23 2001-09-27 Elan Pharmaceuticals, Inc. Compounds and methods to treat alzheimer's disease
GB0008710D0 (en) * 2000-04-07 2000-05-31 Merck Sharp & Dohme Therapeutic compounds
WO2001078721A1 (en) 2000-04-13 2001-10-25 Mayo Foundation For Medical Education And Research Aβ42 LOWERING AGENTS
US6716826B2 (en) * 2000-05-12 2004-04-06 Rodaris Pharmaceuticals Limited Compounds and their uses
US6939857B2 (en) * 2000-05-12 2005-09-06 Rodaris Pharmaceuticals Limited Compounds and their uses
US20040110743A1 (en) 2000-05-19 2004-06-10 Masaomi Miyamato -Secretase inhibitors
GB0015627D0 (en) 2000-06-26 2000-08-16 Rademacher Group Limited Phosphoglycan messengers and their medical uses
PE20020276A1 (en) * 2000-06-30 2002-04-06 Elan Pharm Inc SUBSTITUTE AMINE COMPOUNDS AS ß-SECRETASE INHIBITORS FOR THE TREATMENT OF ALZHEIMER
WO2002002505A2 (en) 2000-06-30 2002-01-10 Elan Pharmaceuticals, Inc. Compounds to treat alzheimer's disease
US6846813B2 (en) * 2000-06-30 2005-01-25 Pharmacia & Upjohn Company Compounds to treat alzheimer's disease
DE10031955A1 (en) 2000-06-30 2002-01-17 Deutsches Krebsforsch Curcumin derivatives with improved water solubility compared to curcumin and medicaments containing them
GB0016681D0 (en) * 2000-07-06 2000-08-23 Merck Sharp & Dohme Therapeutic compounds
US6559891B1 (en) * 2000-08-15 2003-05-06 Sony Corporation Method and apparatus to generate tri-level HDTV synchronization pulses
AU4332302A (en) 2000-11-02 2002-06-24 Us Gov Health & Human Serv Agents useful for reducing amyloid precursor protein and treating demantia and methods of use thereof
CA2427206C (en) 2000-11-02 2011-06-28 Merck Sharp & Dohme Limited Sulfamides as gamma-secretase inhibitors
WO2002055715A1 (en) 2001-01-15 2002-07-18 Hokko Chemical Industry Co., Ltd. Dna encoding d-myo-inositol 1-epimerase
AU2002236988A1 (en) * 2001-02-06 2002-08-19 Novartis Ag Photodynamic therapy of occult choroidal neovascularization linked to age-related macular degeneration
EP1233021A3 (en) * 2001-02-20 2002-11-20 Pfizer Products Inc. An inhibitor of Beta amyloid cleavage enzyme
US20060079533A1 (en) 2001-03-23 2006-04-13 Nieman James A Methods of treating alzheimer's disease
DE10215261A1 (en) * 2001-04-09 2002-10-10 Koenig & Bauer Ag Printing couple of printing machine has single drive motor for transfer cylinder and form cylinder
AU2002256418A1 (en) 2001-04-27 2002-11-11 Vertex Pharmaceuticals Incorporated Inhibitors of bace
US6960664B2 (en) * 2001-05-22 2005-11-01 Pharmacia & Upjohn Company Aza hydroxylated ethyl amine compounds
DE60229846D1 (en) 2001-05-22 2008-12-24 Merck & Co Inc BETA SECRETASE SUBSTRATE AND ITS USE
CA2448834A1 (en) 2001-06-01 2002-12-12 Elan Pharmaceuticals, Inc. Hydroxy alkyl amine derivatives as beta-secretase inhibitors and their use for the treatment of alzheimer's disease and similar diseases
US6684775B2 (en) * 2001-06-07 2004-02-03 Heidelberger Druckmaschinen Ag Printing unit with roll-away inkers
JP2005501015A (en) 2001-06-08 2005-01-13 イーラン ファーマスーティカルズ、インコーポレイテッド How to treat Alzheimer's disease
US7361688B2 (en) 2001-06-11 2008-04-22 Elan Pharmaceuticals, Inc. Substituted aminoalcohols useful in treatment of Alzheimer's disease
JP2004534064A (en) 2001-06-12 2004-11-11 イーラン ファーマスーティカルズ、インコーポレイテッド Macrocyclic molecules useful for treating Alzheimer's disease
JP2005505506A (en) 2001-06-12 2005-02-24 イーラン ファーマスーティカルズ、インコーポレイテッド Macrocycles useful for the treatment of Alzheimer's disease
JP2004532894A (en) * 2001-06-13 2004-10-28 イーラン ファーマスーティカルズ、インコーポレイテッド Amine diol for treating Alzheimer's disease
JP3981597B2 (en) 2001-06-25 2007-09-26 北興化学工業株式会社 Method for producing scyllo-inosose and method for producing scyllo-inositol
CA2451664A1 (en) 2001-06-25 2003-01-03 Elan Pharmaceuticals, Inc. Use of bicyclo compounds for treating alzheimer's disease
MXPA04000140A (en) 2001-06-27 2004-06-03 Elan Pharm Inc Beta-hydroxyamine derivatives useful in treatment of alzheimer's disease.
US7053109B2 (en) 2001-07-10 2006-05-30 Pharmacia & Upjohn Company Aminediols for the treatment of Alzheimer's disease
US6864290B2 (en) 2001-07-10 2005-03-08 Pharmacia & Upjohn, Llp Statine derivatives for the treatment of Alzheimer's disease
EP1406617A1 (en) 2001-07-10 2004-04-14 Elan Pharmaceuticals, Inc. Diaminediols for the treatment of alzheimer's disease
US20030181531A1 (en) * 2003-02-11 2003-09-25 David Sherris Compositions and methods of administering tubulin binding agents for the treatment of ocular diseases
WO2003007944A1 (en) 2001-07-20 2003-01-30 Qlt, Inc. Treatment of macular edema with photodynamic therapy
CN1304390C (en) 2001-08-03 2007-03-14 先灵公司 Novel gamma secretase inhibitors
US7122675B2 (en) * 2001-08-03 2006-10-17 Schering Corporation Gamma secretase inhibitors
BR0212242A (en) 2001-08-28 2004-07-20 Upjohn Co Methods for treating or preventing alzheimer's disease and a disease, treating alzheimer's disease and an individual having or preventing a patient from acquiring a disease or condition, to inhibit beta-secretase activity, the cleavage of a protein isotype. amyloid precursor and the production of amyloid beta peptide in a cell and beta amyloid plaque in an animal to produce a beta-secretase complex, composition and use of a compound
DE10145322A1 (en) * 2001-09-14 2003-04-03 Ina Schaeffler Kg Bearing arrangement for cylinders, rollers or drums
EP1430032A2 (en) 2001-09-24 2004-06-23 Elan Pharmaceuticals, Inc. Substituted amides for the treatment of neurological disorders
MXPA04003245A (en) 2001-10-04 2004-08-11 Elan Pharm Inc Hydroxypropylamines.
AU2002335794A1 (en) 2001-10-05 2003-04-22 Elan Pharmaceuticals, Inc Allylamides useful in the treatment of alzheimer's disease
JP4547152B2 (en) 2001-10-23 2010-09-22 オクラホマ メディカル リサーチ ファウンデーション β-secretase inhibitors and methods of use
US20050038019A1 (en) 2001-10-29 2005-02-17 Beck James P. Hydroxy substituted amides for the treatment of alzheimer's disease
CA2466284A1 (en) 2001-11-08 2003-05-15 Elan Pharmaceuticals, Inc. N, n'-substituted-1,3-diamino-2-hydroxypropane derivatives
OA12812A (en) 2001-11-19 2006-07-10 Elan Pharm Inc (4-phenyl) Piperidin-3-yl-phenylcarboxylate derovatoves and related compounds as beta-secretase inhibitors for the treatment of Alzheimer's Disease.
CA2477585A1 (en) 2001-11-21 2003-06-05 Elan Pharmaceuticals, Inc. Amino acid derivatives useful for the treatment of alzheimer's disease
MXPA04005451A (en) 2001-12-04 2005-01-14 Elan Pharm Inc Peptide isosteres containing a heterocycle useful in the treatment of alzheimer's disease.
JP2005511735A (en) 2001-12-06 2005-04-28 イーラン ファーマスーティカルズ、インコーポレイテッド Substituted hydroxyethylamine
CH698246B1 (en) 2001-12-20 2009-06-30 Hoffmann La Roche Test to identify inhibitors of beta secretases.
US7910586B2 (en) * 2002-01-04 2011-03-22 The Rockefeller University Compositions and methods for prevention and treatment of amyloid-β peptide-related disorders
BR0306724A (en) 2002-01-04 2006-04-11 Elan Pharm Inc amino substituted carboxamides for treatment of alzheimer's disease
AU2003205630A1 (en) 2002-01-18 2003-07-30 The Genetics Company Inc. Beta-secretase inhibitors
US7053220B2 (en) 2002-02-01 2006-05-30 Elan Pharmaceuticals, Inc. Hydroxyalkanoyl aminopyrazoles and related compounds
US20040171614A1 (en) * 2002-02-06 2004-09-02 Schering-Plough Corporation Novel gamma secretase inhibitors
TW200302717A (en) 2002-02-06 2003-08-16 Schering Corp Novel gamma secretase inhibitors
US7256186B2 (en) * 2002-02-06 2007-08-14 Schering Corporation Gamma secretase inhibitors
US20040058313A1 (en) * 2002-04-24 2004-03-25 Abreu Marcio Marc Compositions, targets, methods and devices for the therapy of ocular and periocular disorders
US7585938B2 (en) 2002-04-24 2009-09-08 Hiroshi Mori Gamma-secretase inhibitors
DE60328182D1 (en) * 2002-05-01 2009-08-13 Merck Sharp & Dohme HETEROARYL SUBSTITUTED SPIROCYCLIC SULFAMIDES FOR THE INHIBITION OF GAMMA SECRETASE
AU2003229024A1 (en) 2002-05-07 2003-11-11 Elan Pharmaceuticals, Inc. Succinoyl aminopyrazoles and related compounds
DE10223666B4 (en) * 2002-05-28 2006-06-14 Windmöller & Hölscher Kg Inking system for flexo printing machines
US6723755B2 (en) 2002-06-12 2004-04-20 Piotr Chomczynski Method of treating rosacea
US20060154926A1 (en) 2002-06-11 2006-07-13 Elan Pharmaceuticals, Inc. Methods of treating alzheimer's disease using aryl alkanoic acid amides
WO2003103652A1 (en) 2002-06-11 2003-12-18 Elan Pharmaceuticals, Inc. METHODS OF TREATING ALZHEIMER’S DISEASE USING AROMATICALLY SUBSTITUTED ω-AMINO-ALKANOIC ACID AMIDES AND ALKANOIC ACID DIAMIDES
AU2003247758A1 (en) 2002-06-27 2004-01-19 Elan Pharmaceuticals, Inc. Methods for treating alzheimer's disease using hydroxyethylene compounds containing a heterocyclic amide bond isostere
GB0217909D0 (en) 2002-08-01 2002-09-11 Du Pont Chintzed stretch fabrics
GB0218041D0 (en) 2002-08-02 2002-09-11 Merck Sharp & Dohme Chemical process
US7557137B2 (en) 2002-08-05 2009-07-07 Bristol-Myers Squibb Company Gamma-lactams as beta-secretase inhibitors
PL375747A1 (en) 2002-09-06 2005-12-12 Elan Pharmaceuticals, Inc. 1, 3-diamino-2-hydroxypropane prodrug derivatives
UY27967A1 (en) 2002-09-10 2004-05-31 Pfizer 2-HINDROXI-1,3-DIAMINOALCANE OIL
JP2006501282A (en) 2002-09-27 2006-01-12 イーラン ファーマスーティカルズ、インコーポレイテッド Compounds for the treatment of Alzheimer's disease
AU2003299156B2 (en) 2002-09-30 2008-05-29 Bp Corporation North America Inc. Reduced carbon dioxide emission system and method for providing power for refrigerant compression and electrical power for a light hydrocarbon gas liquefaction process
EP1545582A4 (en) 2002-10-01 2008-09-17 Univ Northwestern Amyloid beta-derived diffusible ligands (addls), addl-surrogates, addl-binding molecules, and uses thereof
GB0223038D0 (en) * 2002-10-04 2002-11-13 Merck Sharp & Dohme Therapeutic compounds
GB0223039D0 (en) 2002-10-04 2002-11-13 Merck Sharp & Dohme Therapeutic compounds
WO2004039759A1 (en) * 2002-10-17 2004-05-13 Sun, Mingjie A natural compound useful for treating diabetes, its preparation and use
CA2507484A1 (en) 2002-11-27 2004-06-17 Elan Pharmaceuticals, Inc. Substituted ureas and carbamates
AU2003279492A1 (en) 2002-12-11 2004-06-30 Pharmacia & Upjohn Company Llc Treatment of diseases with combinations of alpha 7 nicotinic acetylcholine receptor agonists and other compounds
FR2848452B1 (en) 2002-12-12 2007-04-06 Aventis Pharma Sa APPLICATION OF INTESTINAL BILIARY ACID RECAPTURE INHIBITORS FOR THE PREVENTION AND TREATMENT OF ALZHEIMER'S DISEASE
EP1592684B1 (en) 2003-02-04 2008-07-30 F. Hoffmann-La Roche Ag Malonamide derivatives as gamma-secretase inhibitors
EP1603548A4 (en) 2003-02-05 2007-10-10 Myriad Genetics Inc Method and composition for treating neurodegenerative disorders
US7521481B2 (en) 2003-02-27 2009-04-21 Mclaurin Joanne Methods of preventing, treating and diagnosing disorders of protein aggregation
GB0308318D0 (en) 2003-04-10 2003-05-14 Merck Sharp & Dohme Therapeutic agents
WO2004094384A2 (en) 2003-04-21 2004-11-04 Elan Pharmaceuticals, Inc. (hetero) arylamide 2-hydroxy-3-diaminoalkanes for use in the treatment of alzheimer’s disease
CL2004000848A1 (en) 2003-04-21 2005-01-28 Elan Pharmaceuticals Inc Pharm COMPOUNDS DERIVED FROM FENACIL-2-HIDROXI-3-DIAMINOALCANS, INHIBITORS OF THE ENZYME BETASECRETASA, USEFUL TO PREPARE A MEDICINAL PRODUCT TO TREAT ALZHEIMER, DOWN SYNDROME, HEREDITAR CEREBRAL HERIDAL AND OEREDOS
JP2007501859A (en) * 2003-05-13 2007-02-01 シェーリング コーポレイション Bridged n-arylsulfonylpiperidines as γ-secretase inhibitors
KR20060013404A (en) 2003-05-16 2006-02-09 머크 샤프 앤드 돔 리미티드 Cyclic sulfonamides for inhibition of gamma-secretase
US7060698B2 (en) 2003-05-19 2006-06-13 Hoffmann-La Roche Inc. Benzoxazepinone derivatives
CA2528496C (en) 2003-06-05 2011-04-12 Elan Pharmaceuticals, Inc. Acylated amino acid amidyl pyrazoles and related compounds
CA2529994A1 (en) 2003-06-30 2005-01-20 Merck & Co., Inc. N-alkyl phenylcarboxamide beta-secretase inhibitors for the treatment of alzheimer's disease
WO2005004803A2 (en) 2003-07-01 2005-01-20 Merck & Co., Inc. Phenylcarboxylate beta-secretase inhibitors for the treatment of alzheimer's disease
EP1671129A1 (en) 2003-07-21 2006-06-21 Angiogenetics Sweden AB Compounds and methods for promoting angiogenesis by using a gamma-secretase inhibitor or inhibiting the gamma-secretase pathway
GB0318447D0 (en) 2003-08-05 2003-09-10 Merck Sharp & Dohme Therapeutic agents
EP1660443B1 (en) 2003-08-08 2009-03-04 Schering Corporation Cyclic amine bace-1 inhibitors having a benzamide substituent
CN1835936A (en) 2003-08-14 2006-09-20 默克公司 Macrocyclic beta-secretase inhibitors for the treatment of alzheimer's disease
EP1667984B1 (en) 2003-09-24 2011-05-18 Merck Sharp & Dohme Ltd. Gamma-secretase inhibitors
KR100793095B1 (en) 2003-10-01 2008-01-10 주식회사 프로메디텍 Novel Sulfone Amide Derivatives Capable Of Inhibiting BACE
WO2005032471A2 (en) 2003-10-03 2005-04-14 Merck & Co., Inc. Benzylether and benzylamino beta-secretase inhibitors for the treatment of alzheimer's disease
GB0323258D0 (en) * 2003-10-04 2003-11-05 Merck Sharp & Dohme Therapeutic compounds
EP1673347B1 (en) 2003-10-06 2015-08-19 F. Hoffmann-La Roche AG Substituted dibenzo-azepine and benzo-diazepine derivatives useful as gamma-secretase inhibitors
DK1674578T3 (en) 2003-10-14 2010-06-07 Hokko Chem Ind Co Process for the preparation of scyllo-inositol
US20050171112A1 (en) * 2003-11-03 2005-08-04 Probiodrug Ag Combinations useful for the treatment of neuronal disorders
ES2232302B1 (en) 2003-11-07 2006-08-01 Universitat De Les Illes Balears MYO-INOSITOL HEXAFOSFATO FOR TOPICAL USE.
GB0326039D0 (en) * 2003-11-07 2003-12-10 Merck Sharp & Dohme Therapeutic agents
RU2367666C2 (en) 2003-11-11 2009-09-20 Ф.Хоффманн-Ля Рош Аг Phosphinic acid derivatives, beta-secretase inhibitors for treating alzheimer's disease
US7354942B2 (en) 2003-11-24 2008-04-08 Merck & Co., Inc. Benzylether and benzylamino beta-secretase inhibitors for the treatment of Alzheimer's disease
CN1886391A (en) 2003-11-28 2006-12-27 弗·哈夫曼-拉罗切有限公司 Tetronic and tetramic acids as inhibitors of beta-secrease
WO2005065195A2 (en) 2003-12-19 2005-07-21 Merck & Co., Inc. Phenylamide and pyridylamide beta-secretase inhibitors for the treatment of alzheimer's disease
WO2005063796A1 (en) 2003-12-31 2005-07-14 Posco Inhibitors of amyloid precursor protein processing
CN1934251A (en) 2004-01-29 2007-03-21 塞尔卓姆股份公司 Treatment of neurodegenerative diseases by the use of atp7a
PT1713501E (en) 2004-01-29 2008-04-30 Cellzome Ag Treatment of neurodegenerative diseases by the use of gpr49
CA2560773A1 (en) 2004-03-25 2005-10-13 Elan Pharmaceuticals, Inc. 2-amino- and 2-thio-substituted 1,3-diaminopropanes
EP1732906A4 (en) 2004-03-30 2007-11-21 Merck & Co Inc 2-aminothiazole compounds useful as aspartyl protease inhibitors
EP1740570A2 (en) 2004-04-05 2007-01-10 Schering Corporation Novel gamma secretase inhibitors
WO2005103043A1 (en) 2004-04-20 2005-11-03 Merck & Co., Inc. 2, 4, 6-substituted pyridyl derivative compounds useful as beta-secretase inhibitors for the treatment of alzheimer’s disease
EP1740559B1 (en) 2004-04-20 2014-10-15 Merck Sharp & Dohme Corp. 1,3,5-substituted phenyl derivative compounds useful as beta-secretase inhibitors for the treatment of alzheimer's disease
CN1964940A (en) * 2004-05-13 2007-05-16 默克公司 Phenyl carboxamide compounds useful as beta-secretase inhibitors for the treatment of alzheimer's disease
US20060004096A1 (en) * 2004-05-28 2006-01-05 Joseph Larner Method of Treating Endothelial Dysfunction, Oxidative Stress and Related Diseases
JP4406327B2 (en) 2004-06-29 2010-01-27 儀昭 新田 Tire chip manufacturing equipment
US8193250B2 (en) 2004-10-22 2012-06-05 Mount Sinai School Of Medicine Compositions and methods for treating alzheimer's disease and related disorders and promoting a healthy nervous system
ZA200704872B (en) 2004-11-17 2008-12-31 Mclaurin Joanne Compositions comprising scyllo-inositol derivatives and methods to treat disorders of protein aggregation
WO2006088705A1 (en) 2005-02-14 2006-08-24 Wyeth Terphenyl guanidines as [beta symbol] -secretase inhibitors
WO2006112000A1 (en) 2005-03-30 2006-10-26 The Niigata Institute Of Science And Technology Method of synthesizing 2-deoxy-scyllo-inosose by modified strain of e. coli, method of purifying the same and obtained 2-deoxy-scyllo-inosose
KR20060105233A (en) 2005-04-02 2006-10-11 삼성전자주식회사 Hybrid disc and recording and/or reproducing apparatus and method for the same
WO2007002220A2 (en) 2005-06-21 2007-01-04 Bristol-Myers Squibb Company Aminoacetamide acyl guanidines as beta-secretase inhibitors
AU2006270084B2 (en) 2005-07-18 2011-08-25 Merck Sharp & Dohme Corp. Spiropiperidine beta-secretase inhibitors for the treatment of Alzheimer's disease
US20090182021A1 (en) 2005-08-03 2009-07-16 Nanternet Philippe G Tricyclic Beta-Secretase Inhibitors for the Treatment of Alzheimer's Disease
WO2007021793A1 (en) 2005-08-12 2007-02-22 Bristol-Myers Squibb Company Macrocyclic diaminopropanes as beta-secretase inhibitors
JP2009511568A (en) 2005-10-13 2009-03-19 ワラタ ファーマシューティカルズ, インコーポレイテッド Inositol derivatives and their use in the treatment of diseases characterized by abnormal protein folding or aggregation, or amyloid formation, deposition, accumulation or persistence
WO2007050612A1 (en) * 2005-10-25 2007-05-03 Janssen Pharmaceutica N.V. 2-amino-3,4-dihydro-pyrido[3,4-d]pyrimidine derivatives useful as inhibitors of beta-secretase (bace)
US20070197452A1 (en) 2006-02-17 2007-08-23 Mclaurin Joanne Treatment of amyloid-related diseases
CA2579188A1 (en) 2006-02-17 2007-08-17 Joanne Mclaurin Treatment of amyloid-related diseases
EP1993523A4 (en) 2006-02-17 2009-06-10 Joanne Mclaurin Compositions and methods for treatment of disorders of protein aggregation
AU2007222864A1 (en) 2006-03-09 2007-09-13 Joanne Mclaurin A cyclohexane polyalcohol formulation for treatment of disorders of protein aggregation
WO2007134434A1 (en) 2006-05-18 2007-11-29 Merck Frosst Canada Ltd. Phenanthrene derivatives as mpges-1 inhibitors
WO2007134449A1 (en) 2006-05-19 2007-11-29 Waratah Pharmaceuticals Inc. Screening methods for amyloid beta modulators
EP1884531A1 (en) 2006-07-30 2008-02-06 Lonza Compounds GmbH & Co. KG Sheet molding compounds (smc) comprising thermosetting resins based on renewable resources
CA2697675A1 (en) 2006-08-25 2008-02-28 Allan Basbaum Intrathecal administration of triptan compositions to treat non-migraine pain
JP2010502395A (en) 2006-09-06 2010-01-28 エーオーテックス, インコーポレイテッド Prosthetic heart valve, implantation system and method
US20100173960A1 (en) 2006-09-21 2010-07-08 Antonio Cruz The Combination of a Cyclohexanehexol and a NSAID for the Treatment of Neurodegenerative Diseases
WO2008061373A1 (en) 2006-11-24 2008-05-29 Waratah Pharmaceuticals Inc. Combination treatments for alzheimer's disease and similar diseases
WO2008082658A2 (en) 2006-12-29 2008-07-10 Normoxys, Inc. Cyclitols and their derivatives and their therapeutic applications
WO2008098371A1 (en) 2007-02-16 2008-08-21 The Royal Institution For The Advancement Of Learning/Mcgill University Compositions and method for reducing amyloid beta in a mammal
WO2008124931A1 (en) 2007-04-12 2008-10-23 Joanne Mclaurin Use of cyclohexanehexol derivatives in the treatment of amyotrophic lateral sclerosis
US20100331267A1 (en) 2007-04-12 2010-12-30 Mclaurin Joanne Use of cyclohexanehexol derivatives in the treatment of alpha-synucleinopathies
SI2148667T1 (en) 2007-04-12 2013-12-31 Waratah Pharmaceuticals, Inc. Use of cyclohexanehexol derivatives in the treatment of ocular diseases
CA2683548A1 (en) 2007-04-12 2008-10-23 Joanne Mclaurin Use of cyclohexanehexol derivatives for the treatment of polyglutamine diseases
JP5034916B2 (en) 2007-12-10 2012-09-26 富士通セミコンダクター株式会社 Performance evaluation model generation method, system performance evaluation method, and performance evaluation model generation apparatus
WO2010017297A2 (en) 2008-08-06 2010-02-11 Invista Technologies S.A.R.L. Preparation of elastic composite structures useful for components of disposable hygiene products and articles of apparel
ES2521674T3 (en) * 2008-09-15 2014-11-13 Transition Therapeutics Ireland Limited Methods of treatment of hyperuricemia and associated pathological conditions
WO2010040232A1 (en) 2008-10-09 2010-04-15 Waratah Pharmaceuticals Inc. Use of scyllo-inositols for the treatment of macular degeneration-related disorders
WO2012173808A1 (en) 2011-06-03 2012-12-20 Elan Pharmaceuticals, Inc. Scyllo-inositol for the treatment of behavioral and psychiatric disorders

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4758430A (en) * 1987-01-21 1988-07-19 Robert Sabin Method of treatment of Alzheimer's disease using phytic acid
US4847082A (en) * 1987-01-21 1989-07-11 Robert Sabin Method of treatment of Alzheimer's disease using phytic acid
US5112814A (en) * 1990-10-24 1992-05-12 Robert Sabin Method of treatment of Parkinson's disease using phytic acid

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
MCLAURIN J ET AL: "Review: Modulating factors in amyloid-beta fibril formation", JOURNAL OF STRUCTURAL BIOLOGY, vol. 130, no. 2-3, June 2000 (2000-06-01), pages 259 - 270, XP002280181, ISSN: 1047-8477 *
MCLAURIN JOANNE ET AL: "Inositol stereoisomers stabilize an oligomeric aggregate of Alzheimer amyloid beta peptide and inhibit Abeta-induced toxicity", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 275, no. 24, 16 June 2000 (2000-06-16), pages 18495 - 18502, XP002280180, ISSN: 0021-9258 *

Cited By (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8859628B2 (en) 2003-02-27 2014-10-14 JoAnne McLaurin Method for preventing, treating and diagnosing disorders of protein aggregation
US9833420B2 (en) 2003-02-27 2017-12-05 JoAnne McLaurin Methods of preventing, treating, and diagnosing disorders of protein aggregation
US10695449B2 (en) 2004-02-24 2020-06-30 The General Hospital Corporation Catalytic radiofluorination
US10058625B2 (en) 2004-02-24 2018-08-28 The General Hospital Corporation Catalytic radiofluorination
EP2186518A1 (en) * 2004-11-17 2010-05-19 McLaurin, Joanne Composition and methods for treatment of disorders of protein aggregation
CN103301094A (en) * 2004-11-17 2013-09-18 乔安妮·麦克劳林 Compositions comprising scyllo-inositol derivatives and methods to treat disorders of protein aggregation
AU2005306531B2 (en) * 2004-11-17 2012-02-23 Joanne Mclaurin Compositions comprising scyllo-inositol derivatives and methods to treat disorders of protein aggregation
JP2008520589A (en) * 2004-11-17 2008-06-19 ジョアン マクローリン, Compositions comprising sylinositol derivatives and methods for treating protein aggregation disorders
WO2006053428A1 (en) * 2004-11-17 2006-05-26 Joanne Mclaurin Compositions comprising scyllo-inositol derivatives and methods to treat disorders of protein aggregation
WO2007119108A2 (en) 2005-10-13 2007-10-25 Waratah Pharmaceuticals Inc. Inositol derivatives and their uses in the treatment of diseases characterized by abnormal protein folding or aggregation or amyloid formation, deposition, accumulation or persistence
JP2009511568A (en) * 2005-10-13 2009-03-19 ワラタ ファーマシューティカルズ, インコーポレイテッド Inositol derivatives and their use in the treatment of diseases characterized by abnormal protein folding or aggregation, or amyloid formation, deposition, accumulation or persistence
WO2007041855A1 (en) * 2005-10-13 2007-04-19 Waratah Pharmaceuticals, Inc. Scyllo-inositol derivatives and their use in the treatment of diseases characterised by abnormal protein folding or aggregation or amyloid formation, deposition, accumulation or persistence
WO2007119108A3 (en) * 2005-10-13 2008-03-13 Waratah Pharmaceuticals Inc Inositol derivatives and their uses in the treatment of diseases characterized by abnormal protein folding or aggregation or amyloid formation, deposition, accumulation or persistence
US7807175B2 (en) * 2005-12-12 2010-10-05 Ac Immune Sa Therapeutic vaccine
US8603487B2 (en) 2005-12-12 2013-12-10 Ac Immune S.A. Palmitoylated AB 1-15 in a liposome as a treatment for amyloid associated diseases
WO2007129221A3 (en) * 2006-02-17 2008-03-20 Joanne Mclaurin Compositions comprising an epi-inositol compounds and methods for treatment of disorders of protein aggregation
JP2009529502A (en) * 2006-03-09 2009-08-20 ワラタ ファーマシューティカルズ, インコーポレイテッド Cyclohexane polyhydric alcohol formulations for the treatment of protein aggregation disorders
WO2007101353A1 (en) * 2006-03-09 2007-09-13 Waratah Pharmaceuticals Inc. A cyclohexane polyalcohol formulation for treatment of disorders of protein aggregation
WO2008034244A1 (en) * 2006-09-21 2008-03-27 Waratah Pharmaceuticals Inc. The combination of a cyclohexanehexol and a nsaid for the treatment of neurodegenerative diseases
US20100292157A1 (en) * 2006-11-24 2010-11-18 Antonio Cruz Combination Treatments for Alzheimer's Disease and Similar Diseases
US20100331267A1 (en) * 2007-04-12 2010-12-30 Mclaurin Joanne Use of cyclohexanehexol derivatives in the treatment of alpha-synucleinopathies
WO2008124930A1 (en) * 2007-04-12 2008-10-23 Joanne Mclaurin Use of cyclohexanehexol derivatives for the treatment of polyglutamine diseases
US10857247B2 (en) 2008-03-21 2020-12-08 The General Hospital Corporation Compounds and compositions for the detection and treatment of Alzheimer's disease and related disorders
EP3231804A1 (en) * 2008-03-21 2017-10-18 The General Hospital Corporation Compounds and compositions for the detection and treatment of alzheimer's disease and related disorders
WO2010011012A1 (en) 2008-07-22 2010-01-28 Postech Academy-Industry Foundation Inositol and trehalose derivatives and pharmaceutical compositions for treating neurodegenerative diseases comprising the same
US9371350B2 (en) 2008-07-22 2016-06-21 Postech Foundation Trehalose derivatives and pharmaceutical compositions for treating neurodegenerative diseases
WO2010063701A2 (en) * 2008-12-02 2010-06-10 Ge Healthcare Limited In vivo imaging method
WO2010063701A3 (en) * 2008-12-02 2010-08-05 Ge Healthcare Limited In vivo imaging method for parkinson' s disease
WO2012051395A1 (en) 2010-10-13 2012-04-19 Elan Pharmaceuticals, Inc. Methods of synthesis of scyllitol and related compounds
US8889921B2 (en) 2010-10-13 2014-11-18 Transition Therapeutics Ireland Limited Methods of synthesis of scyllitol and related compounds
US9116157B2 (en) 2010-11-05 2015-08-25 Brandeis University Ice-cleaved alpha-synuclein as a biomarker
WO2012173808A1 (en) * 2011-06-03 2012-12-20 Elan Pharmaceuticals, Inc. Scyllo-inositol for the treatment of behavioral and psychiatric disorders
CN103906520A (en) * 2011-06-03 2014-07-02 伊兰制药公司 Scyllo-inositol for the treatment of behavioral and psychiatric disorders
WO2016047823A1 (en) * 2014-09-25 2016-03-31 영남대학교 산학협력단 Composition for inhibiting cellular senescence containing melandrium firmum rohrbach extract or bornesitol separated from same as active ingredient
US11844846B2 (en) 2018-05-16 2023-12-19 Emory University Styrylbenzothiazole derivatives and uses in imaging methods

Also Published As

Publication number Publication date
EP2823813A1 (en) 2015-01-14
US7521481B2 (en) 2009-04-21
PT1608350E (en) 2009-08-28
JP4999453B2 (en) 2012-08-15
AU2004216544A1 (en) 2004-09-10
SI2311445T1 (en) 2014-08-29
CA2516563A1 (en) 2004-09-10
EP2311445B1 (en) 2014-03-26
IL220065A (en) 2015-08-31
BRPI0407910A (en) 2006-02-14
AU2009251035A1 (en) 2010-01-14
JP2016117743A (en) 2016-06-30
IL220065A0 (en) 2012-09-24
DK2153829T3 (en) 2014-10-27
US9833420B2 (en) 2017-12-05
US20140073701A1 (en) 2014-03-13
DE602004021362D1 (en) 2009-07-16
US20120157549A1 (en) 2012-06-21
US8859628B2 (en) 2014-10-14
US20040204387A1 (en) 2004-10-14
ES2327521T3 (en) 2009-10-30
CY1115732T1 (en) 2017-01-25
CA2516563C (en) 2013-08-20
SI1608350T1 (en) 2009-12-31
PT2311445E (en) 2014-06-25
JP2006522740A (en) 2006-10-05
EP2153829A1 (en) 2010-02-17
DK2311445T3 (en) 2014-06-16
US20070078099A1 (en) 2007-04-05
CY1109338T1 (en) 2014-07-02
DK1608350T3 (en) 2009-09-07
US20090227686A1 (en) 2009-09-10
EP1608350B1 (en) 2009-06-03
AU2009251035B2 (en) 2012-12-13
IL170476A (en) 2013-05-30
US20150164821A1 (en) 2015-06-18
JP2014065731A (en) 2014-04-17
EP2311445A1 (en) 2011-04-20
EP2153829B1 (en) 2014-08-13
AU2004216544B2 (en) 2009-09-24
US20150031776A1 (en) 2015-01-29
ES2475977T3 (en) 2014-07-11
ATE432694T1 (en) 2009-06-15
EP1608350A1 (en) 2005-12-28
PT2153829E (en) 2014-10-16
JP2011105733A (en) 2011-06-02
ES2520047T3 (en) 2014-11-11
SI2153829T1 (en) 2014-12-31

Similar Documents

Publication Publication Date Title
US9833420B2 (en) Methods of preventing, treating, and diagnosing disorders of protein aggregation
Spangenberg et al. Sustained microglial depletion with CSF1R inhibitor impairs parenchymal plaque development in an Alzheimer’s disease model
Núñez et al. Iron toxicity in neurodegeneration
Frost et al. Pyroglutamate-3 amyloid-β deposition in the brains of humans, non-human primates, canines, and Alzheimer disease–like transgenic mouse models
Dorninger et al. Disturbed neurotransmitter homeostasis in ether lipid deficiency
Wildburger et al. Amyloid-β plaques in clinical Alzheimer’s disease brain incorporate stable isotope tracer in vivo and exhibit nanoscale heterogeneity
Sawa et al. Impact of increased APP gene dose in Down syndrome and the Dp16 mouse model
Ben Khedher et al. Apolipoprotein E4–driven effects on inflammatory and neurotrophic factors in peripheral extracellular vesicles from cognitively impaired, no dementia participants who converted to Alzheimer's disease
Dehelean et al. Trends in glycolipid biomarker discovery in neurodegenerative disorders by mass spectrometry
Zhang et al. Ageing related thyroid deficiency increases brain-targeted transport of liver-derived ApoE4-laden exosomes leading to cognitive impairment
Hedera et al. Inherited dementias
Figueiredo et al. Drug repurposing of dopaminergic drugs to inhibit Ataxin-3 aggregation
Lei The roles of sphingosine kinase 2 in behaviour, myelination and protection against Alzheimer’s disease
CN101385722A (en) Method of preventing, treating and diagnosing disorders of protein aggregation
Huber Proteostasis & Neuroinflammation in Alzheimer's Disease
Dobson Development of a Cell Culture Model to Investigate Aluminium's Neurotoxicity, and Putative Role in Alzheimer's Disease
Nykänen Longitudinal Synaptic, Inflammatory, and Dopaminergic System Assessment in a SynFib Rat
McRae et al. Biomarkers in Alzheimer‟ s Disease
Ali-Rahmani HFE Gene Variants and Dysregulation of Cholesterol and Iron Metabolism in Alzheimer Disease and Cancer

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DPEN Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed from 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2004216544

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2516563

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 170476

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2006501433

Country of ref document: JP

ENP Entry into the national phase

Ref document number: 2004216544

Country of ref document: AU

Date of ref document: 20040227

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2004216544

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2004715226

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 20048113358

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2004715226

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0407910

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: 2007078099

Country of ref document: US

Ref document number: 10547286

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 10547286

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 220065

Country of ref document: IL